EP3371190B1 - Heterocyclic compounds as pi3k-gamma inhibitors - Google Patents

Heterocyclic compounds as pi3k-gamma inhibitors Download PDF

Info

Publication number
EP3371190B1
EP3371190B1 EP16805238.9A EP16805238A EP3371190B1 EP 3371190 B1 EP3371190 B1 EP 3371190B1 EP 16805238 A EP16805238 A EP 16805238A EP 3371190 B1 EP3371190 B1 EP 3371190B1
Authority
EP
European Patent Office
Prior art keywords
alkyl
pyrazolo
amino
pyrimidine
chloro
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP16805238.9A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP3371190A1 (en
Inventor
Artem SHVARTSBART
Stacey Shepard
Andrew P. Combs
Lixin Shao
Nikoo Falahatpisheh
Ge Zou
Andrew W. Buesking
Richard B. Sparks
Eddy W. Yue
Ravi Jalluri
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Incyte Corp
Original Assignee
Incyte Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Incyte Corp filed Critical Incyte Corp
Priority to RS20220521A priority Critical patent/RS63359B1/sr
Priority to HRP20220599TT priority patent/HRP20220599T1/hr
Priority to EP22162035.4A priority patent/EP4086259A1/en
Priority to SI201631549T priority patent/SI3371190T1/sl
Publication of EP3371190A1 publication Critical patent/EP3371190A1/en
Application granted granted Critical
Publication of EP3371190B1 publication Critical patent/EP3371190B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention provides heterocyclic compounds that modulate the activity of phosphoinositide 3-kinases-gamma (PI3K ⁇ ) and are useful in the treatment of diseases related to the activity of PI3K ⁇ including, for example, autoimmune diseases, cancer, cardiovascular diseases, and neurodegenerative diseases.
  • diseases related to the activity of PI3K ⁇ including, for example, autoimmune diseases, cancer, cardiovascular diseases, and neurodegenerative diseases.
  • PI3Ks The phosphoinositide 3-kinases (PI3Ks) belong to a large family of lipid signaling kinases that phosphorylate phosphoinositides at the D3 position of the inositol ring ( Cantley, Science, 2002, 296(5573):1655-7 ). PI3Ks are divided into three classes (class I, II, and III) according to their structure, regulation and substrate specificity.
  • Class I PI3Ks which include PI3K ⁇ , PI3K ⁇ , PI3K ⁇ , and PI3K ⁇ , are a family of dual specificity lipid and protein kinases that catalyze the phosphorylation of phosphatidylinosito-4,5-bisphosphate (PIP 2 ) giving rise to phosphatidylinosito-3,4,5-trisphosphate (PIP 3 ).
  • PIP 3 functions as a second messenger that controls a number of cellular processes, including growth, survival, adhesion and migration.
  • PI3K isoforms exist as heterodimers composed of a catalytic subunit (p110) and a tightly associated regulatory subunit that controls their expression, activation, and subcellular localization.
  • PI3K ⁇ , PI3K ⁇ , and PI3K ⁇ associate with a regulatory subunit known as p85 and are activated by growth factors and cytokines through a tyrosine kinase-dependent mechanism ( Jimenez, et al., J Biol Chem., 2002, 277(44):41556-62 ) whereas PI3K ⁇ associates with two regulatory subunits (p101 and p84) and its activation is driven by the activation of G-protein-coupled receptors ( Brock, et al., J Cell Biol., 2003, 160(1):89-99 ).
  • PI3K ⁇ and PI3K ⁇ are ubiquitously expressed. In contrast, PI3K ⁇ and PI3K ⁇ are predominantly expressed in leukocytes ( Vanhaesebroeck, et al., Trends Biochem Sci., 2005, 30(4): 194-204 ).
  • PI3K ⁇ is mainly restricted to hematopoietic system, although it can be also detected at lower level in endothelium, heart and brain.
  • PI3K ⁇ knock-out or kinase dead knock in mice are normal and fertile and do not present any overt adverse phenotypes. Analysis at the cellular level indicates that PI3K ⁇ is required for GPCR ligand-induced PtdINs (3,4,5)P3 production, chemotaxis and respiratory burst in neutrophils.
  • PI3K ⁇ -null macrophages and dendritic cell exhibit reduced migration towards various chemoattractants. T-cells deficient in PI3K ⁇ show impaired cytokine production in response to anti-CD3 or Con A stimulation.
  • PI3K ⁇ working downstream of adenosine A3A receptor is critical for sustained degranulation of mast cells induced by FC ⁇ RI crosslinking with IgE.
  • PI3K ⁇ is also essential for survival of eosinophils ( Ruckle et al., Nat. Rev. Drug Discovery, 2006, 5, 903-918 )
  • PI3K ⁇ was shown to be important for recruitment of neutrophils and eosinopohils to airways and degranulation of mast cells (see e.g. Laffargue et al., Immunity, 2002, 16, 441-451 ; Prete et al., The EMBO Journal, 2004, 23, 3505-3515 ; Pinho et al., L.
  • PI3K ⁇ -/- mice were largely protected in four different models of rheumatoid arthritis (CIA, ⁇ -CII-IA, K/BxN serum transfer and TNF transgenic) and PI3K ⁇ inhibition suppressed the progression of joint inflammation and damage in the CIA and ⁇ -CII-IA models (see e.g., Camps et al., Nat.
  • AD Alzheimer's disease
  • PI3K ⁇ inhibition was shown to attenuate A ⁇ (1-40)-induced accumulation of activated astrocytes and microglia in the hippocampus and prevent the peptide-induced congnitive deficits and synaptic dysfunction in a mouse model of AD ( Passos et al., Brain Behav. Immun.
  • PI3K ⁇ deficiency or inhibition also was shown to delay onset and alleviate symptoms in experimental autoimmune encephalomyelitis in mice, a mouse model of human multiple sclerosis, which is another form of neurodegeneration disease (see e.g., Rodrigues et al., J. Neuroimmunol. 2010, 222, 90-94 ; Berod et al., Euro. J. Immunol. 2011, 41, 833-844 ; Comerford et al., PLOS one, 2012, 7, e45095 ; Li et al., Neuroscience, 2013, 253, 89-99 ).
  • PI3K ⁇ is reported to mediate various inflammatory processes, its role as an immune oncology target has also been investigated.
  • a recent study reported that PI3K ⁇ deficiency suppressed tumor growth in the syngeneic models of lung cancer, pancreatic cancer and melanoma (LLC, PAN02 and B16).
  • PI3K ⁇ deficiency or inhibition also inhibited tumor growth in a spontaneous breast cancer model ( Schmid et al., Cancer Cell, 2011, 19, 715-727 ).
  • M2 macrophages promote tumor growth and progression by secreting immunosuppressive factors such arginase 1, which depletes the tumor microenvironment of arginine, thereby promoting T-cell death and NK cell inhibition ( Schmidt et al., Cancer Res. 2012, 72 (Suppl 1: Abstract, 411 ; Kaneda et al., Cancer Res., 74 (Suppl 19: Abstact 3650 )).
  • PI3K ⁇ may play a direct role in cancer cells.
  • PI3K ⁇ is reported to be required for signaling from the Kaposi's sarcoma-associated herpevirus encoded ⁇ GPCR oncogene and tumor growth in a mouse model of sarcoma ( Martin et al., Cancer Cell, 2011, 19, 805-813 ).
  • PI3K ⁇ was also suggested to be required for growth of T-ALL ( Subramanjam et al., Cancer Cell, 2012, 21, 459-472 ), PDAC and HCC cells ( Falasca and Maffucci, Frontiers in Physiology, 2014, 5, 1-10 ).
  • PI3K ⁇ gene was found to contain second highest scoring predicted driven mutation (R839C) among the set of genes not previously identified as a driver in pancreatic cancer ( Carter et al., Cancer Biol. Ther. 2010, 10, 582-587 ).
  • PI3K ⁇ deficiency also has been reported to offer protection to experimental animals in different cardiovascular disease models.
  • lack of PI3K ⁇ would reduce angiotension-evoked smooth muscle contraction and, therefore, protect mice from angiotension-induced hypertension ( Vecchione et al., J. Exp. Med. 2005, 201, 1217-1228 ).
  • PI3K ⁇ inhibition provided potent cardioprotection, reducing infarct development and preserving myocardial function ( Doukas et al., Proc. Natl. Acad. Sci. USA, 2006, 103, 19866-19871 ).
  • WO 2013/154878 and WO 2015/051241 describe some heterocyclic compounds which are inhibitors of PI3K ⁇ and PI3K ⁇ .
  • the present invention related to, inter alia, compounds of Formula (II): or pharmaceutically acceptable salts, wherein constituent members are defined herein.
  • the present invention further provides pharmaceutical compositions comprising a compound of Formula II, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the present invention further provides compounds of Formula II, and pharmaceutically acceptable salts thereof, for use in methods of treating a disease or a disorder associated with abnormal PI3K ⁇ kinase expression or activity in a patient by administering to a patient a therapeutically effective amount of a compound of Formula II, or a pharmaceutically acceptable salt thereof.
  • the present invention further provides a compound of Formula II, or a pharmaceutically acceptable salt thereof, for use in treating a disorder or disease as defined in the claims.
  • the present invention further provides use of a compound of Formula II, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for use in any of the methods described herein.
  • the present invention provides, inter alia, a compound of Formula II: or a pharmaceutically acceptable salt thereof; wherein:
  • R 7 , R 8 , and R 9 are each independently H, halo, CN, -OH, -C(O)O(C 1-4 alkyl), -C(O)NH 2 , - C(O)NH(C 1-4 alkyl), -C(O)N(C 1-4 alkyl) 2 , (C 1-4 alkyl)C(O)NH-, (C 1-4 alkyl)C(O)-, C 1-4 alkylthio, - NH(C 1-4 alkyl), -N(C 1-4 alkyl) 2 , C 1-4 alkyl, C 1-4 alkoxy, C 1-4 haloalkyl, C 1-4 haloalkoxy, -SO 2 (C 1-4 alkyl), -SO 2 NH(C 1-4 alkyl), -SO 2 N(C 1-4 alkyl) 2 , and (C 1-4 alkyl)SO 2 NH-, wherein the -C(O)O
  • X 1 is C; and X 2 is C.
  • X 1 is N; and X 2 is C.
  • X 1 is C; and X 2 is N.
  • X 3 is N.
  • X 3 is NR 3a .
  • X 3 is CR 3 .
  • X 4 is N.
  • X 4 is NR 4a .
  • X 4 is CR 4 .
  • X 5 is N.
  • X 5 is NR 5a .
  • X 5 is CR 5 .
  • W is CH.
  • W is N.
  • W is CH; X 1 is C; X 2 is N; X 3 is CR 3 ; X 4 is N; and X 5 is CR 5 .
  • W is CH; X 1 is C; X 2 is C; X 3 is NR 3a ; X 4 is N; and X 5 is CR 5 .
  • W is CH; X 1 is C; X 2 is C; X 3 is N; X 4 is NR 4a ; and X 5 is CR 5 .
  • R 1 is H, C 3-6 cycloalkyl, C 1-6 haloalkyl or C 1-6 alkyl.
  • R 1 is H or C 1-6 alkyl.
  • R 1 is C 1-6 alkyl.
  • R 1 is methyl, ethyl or 1-propyl.
  • R 1 is methyl
  • R 2 is OR 13 , C 6-10 aryl, 5-10 memberd heterocycloalkyl, or 5-10 membered heteroaryl; wherein said C 6-10 aryl, 5-10 memberd heterocycloalkyl, and 5-10 membered heteroaryl of R 2 are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R j substituents.
  • R 2 is OR 13 , C 6-10 aryl, or 5-10 membered heteroaryl; wherein said C 6-10 aryl or 5-10 membered heteroaryl of R 2 are each optionally substituted with 1, 2, 3, 4, or 5 independently selected R j substituents.
  • R 2 is OR 13 or C 6-10 aryl; wherein said C 6-10 aryl of R 2 is optionally substituted with 1, 2, or 3 independently selected R j substituents.
  • R 2 is C 1-6 alkoxy, phenyl, or monocyclic 5-6 membered heteroaryl; wherein said C 6-10 aryl or monocyclic 5-6 membered heteroaryl of R 2 are each optionally substituted with 1, 2, or 3 independently selected R j substituents.
  • R 2 is C 1-6 alkoxy or phenyl, wherein said phenyl of R 2 is optionally substituted with 1, 2, or 3 independently selected R j substituents.
  • R 2 is C 1-6 alkoxy, phenyl, 5-6 membered heterocycloalkyl, or monocyclic 5-6 membered heteroaryl; wherein said C 6-10 aryl, 5-6 membered heterocycloalkyl, monocyclic 5-6 membered heteroaryl of R 2 are each optionally substituted with 1, 2, or 3 independently selected R j substituents.
  • R 2 is 5-6 membered heterocycloalkyl which is optionally substituted with 1, 2, or 3 independently selected R j substituents.
  • R 2 is 1,1-dioxidothiomorpholin-4-yl optionally substituted with 1 or 2 independently selected R j substituents.
  • R 2 is ethoxy, 1,1-dioxidothiomorpholino optionally substituted with 1 or 2 C 1-4 independently selected alkyl substituents, or phenyl optionally substituted with halo.
  • R 2 is ethoxy or phenyl optionally substituted with halo.
  • R 2 is 1,1-dioxidothiomorpholino optionally substituted with 1 or 2 independently selected C 1-4 alkyl substituents.
  • R 2 is 2-methyl-1,1-dioxidothiomorpholino.
  • each R j is independently selected from halo, CN, C 1-4 alkyl, C 1-4 haloalkyl, C 2-4 alkenyl, C 2-4 alkynyl, NHOR k , OR k , C(O)R k , C(O)NR k R k , C(O)OR k , OC(O)R k , OC(O)NR k R k , NHR k , NR k R k , NR k C(O)R k , S(O) 2 R k , NR k S(O) 2 R k , and S(O) 2 NR k R k .
  • each R k is independently selected from H, C 1-4 alkyl, and C 1-4 haloalkyl.
  • each R j is independently selected from halo, CN, C 1-4 alkyl, C 1-4 haloalkyl, C 1-4 alkoxy, C 1-4 haloalkoxy, amino, C 1-4 alkylamino, and di(C 1-4 alkyl)amino.
  • each R j is independently C 1-4 alkyl or halo.
  • each R j is independently C 1-4 alkyl.
  • each R j is independently methyl.
  • each R j is independently halo.
  • R 2 is ethoxy, phenyl, or 3-fluorophenyl.
  • R 3a is selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, C 6-10 aryl, C 3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10 cycloalkyl-C 1-4 alkyl-, (5-10 membered heteroaryl)-C 1-4 alkyl-, (4-10 membered heterocycloalkyl)-C 1-4 alkyl-; wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, C 6-10 aryl, C 3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10 cycloalkyl;
  • R 3a is H, C 1-6 alkyl, C 2-6 alkynyl, C 3-7 cycloalkyl, phenyl-C 1-4 alkyl-, (5-6 membered heteroaryl)-C 1-4 alkyl, and (4-6 membered heterocycloalkyl)-C 1-4 alkyl; wherein said C 1-6 alkyl, C 2-6 alkynyl, C 3-7 cycloalkyl, phenyl-C 1-4 alkyl-, (5-6 membered heteroaryl)-C 1-4 alkyl, and (4-6 membered heterocycloalkyl)-C 1-4 alkyl of R 3a are each optionally substituted by 1, 2, 3, or 4 independently selected R b substituents.
  • each R b is independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, CN, OH, NH 2 , NHOR c , OR c , C(O)R c , C(O)NR c R c , C(O)OR c , OC(O)R c , OC(O)NR c R c , NHR c , NR c R c , NR c C(O)R c , NR c C(O)OR c , NR c S(O)R c , NR c S(O) 2 R c , S(O) 2 R c and S(O) 2 NR c R c .
  • each R b is independently selected from C 1-4 alkyl, OH, OR c , CN, C(O)NR c R c , and NR c R c .
  • each R c is independently selected from H and C 1-6 alkyl.
  • R 3a is H, methyl, isopropyl, isobutyl, -CH 2 C ⁇ CCH 3 , -CH 2 CH 2 OH, - CH 2 CH 2 OCH 3 , -CH 2 CN, -CH 2 CH 2 NH 2 , -CH 2 C(O)NH 2 , benzyl, cyclobutyl, -CH 2 -(1-methyl-1H-pyrazol-3-yl), or -CH 2 CH 2 -(morpholin-4-yl).
  • R 3 is H, halo, CN, C 1-6 alkyl, or C 1-6 haloalkyl.
  • R 3 is H or C 1-6 alkyl.
  • R 3 is H, methyl, or ethyl.
  • R 4a is selected from H, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, C 6-10 aryl, C 3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10 cycloalkyl-C 1-4 alkyl-, (5-10 membered heteroaryl)-C 1-4 alkyl-, (4-10 membered heterocycloalkyl)-C 1-4 alkyl-; wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, C 6-10 aryl, C 3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10 cycloalkyl;
  • R 4a is selected from H, C 1-6 alkyl, C 2-6 alkynyl, C 3-10 cycloalkyl, phenyl-C 1-4 alkyl-, and (5-6 membered heteroaryl)-C 1-4 alkyl-, wherein said C 1-6 alkyl, C 2-6 alkynyl, C 3-10 cycloalkyl, phenyl-C 1-4 alkyl-, and (5-6 membered heteroaryl)-C 1-4 alkyl- of R 4a are each optionally substituted by 1, 2, 3, or 4 independently selected R b substituents.
  • R 4a is selected from H, C 1-6 alkyl, C 2-6 alkynyl, C 3-10 cycloalkyl, C 6-10 aryl-C 1-4 alkyl-, and (5-10 membered heteroaryl)-C 1-4 alkyl-, wherein said C 1-6 alkyl and (4-10 membered heteroaryl)-C 1-4 alkyl- of R 4a are each optionally substituted by 1, 2, 3, or 4 independently selected R b substituents.
  • each R b is independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl, CN, OH, NH 2 , NHOR c , OR c , C(O)R c , C(O)NR c R c , C(O)OR c , OC(O)R c , OC(O)NR c R c , NHR c , NR c R c , NR c C(O)R c , NR c C(O)OR c , NR c S(O)R c , NR c S(O) 2 R c , S(O) 2 R c and S(O) 2 NR c R c .
  • each R b is independently selected from C 1-4 alkyl, OH, OR c , CN, NR c R c , and C(O)NR c R c .
  • each R c is independently H or C 1-6 alkyl; or any two R c substituents together with the nitrogen atom to which they are attached form a 4-, 5-, 6-, or 7-membered heterocycloalkyl group optionally substituted with 1, 2, or 3 independently selected R h substituents.
  • R 4a is H, methyl, ethyl, isopropyl, isobutyl, -CH 2 C ⁇ CCH 3 , - CH 2 CH 2 OH, -CH 2 CH 2 OCH 3 , -CH 2 CN, -CH 2 CH 2 NH 2 , cyclobutyl, benzyl, -CH 2 C(O)NH 2 , -CH 2 -(1-methyl-1H-pyrazol-3-yl), or -CH 2 C(O)-(morpholin-4-yl).
  • R 4 is selected from H, halo, CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, C 1-6 haloalkoxy, C 6-10 aryl, C 3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl, C 6-10 aryl-C 1-4 alkyl-, C 3-10 cycloalkyl-C 1-4 alkyl-, (5-10 membered heteroaryl)-C 1-4 alkyl-, (4-10 membered heterocycloalkyl)-C 1-4 alkyl-; wherein the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 haloalkyl, C 1-6 haloalkoxy, C 6-10 aryl, C 3-10 cycloalkyl, 5-10 membered heteroaryl, 4-10 membered heterocycloalkyl,
  • R 4 is selected from H, halo, CN, C 1-6 alkyl, C 2-6 alkynyl, C 3-10 cycloalkyl, phenyl-C 1-4 alkyl-, and (5-6 membered heteroaryl)-C 1-4 alkyl-, wherein said C 1-6 alkyl, C 1-6 alkynyl, C 3-10 cycloalkyl, phenyl-C 1-4 alkyl-, and (5-6 membered heteroaryl)-C 1-4 alkyl- of R 4 are each optionally substituted by 1, 2, 3, or 4 independently selected R b substituents.
  • R 4 is H, halo, CN, C 1-6 alkyl, or C 1-6 haloalkyl.
  • R 4 is H or C 1-6 alkyl.
  • R 5 is H, halo, C 1-6 alkyl, or C 1-6 haloalkyl.
  • R 5 is H, halo, or C 1-6 alkyl.
  • R 5 is H, bromo, chloro, fluoro, methyl, ethyl, n-propyl, or isopropyl. In some embodiments, R 5 is H, bromo, or methyl.
  • R 6 is H, halo, CN, C 1-6 alkyl, or C 1-6 haloalkyl.
  • R 6 is halo, CN, or C 1-6 alkyl.
  • R 6 is selected from chloro, CN, and methyl.
  • R 7 is H, 4-10 membered heterocycloalkyl, or 5-10 membered heteroaryl, wherein the 4-10 membered heterocycloalkyl and 5-10 membered heteroaryl are each optionally substituted with 1 or 2 independently selected R q substituents.
  • R 7 is H, 5-6 membered heterocycloalkyl, or 5-6 membered heteroaryl, wherein the 5-6 membered heterocycloalkyl and 5-6 membered heteroaryl are each optionally substituted with 1 or 2 independently selected R q substituents.
  • R 7 is H, piperidinyl, or pyridyl, wherein the piperidinyl and pyridyl groups are each optionally substituted with 1 or 2 independently selected R q substituents.
  • R 7 is H, piperidinyl, or pyridyl, wherein the piperidinyl and pyridyl groups are each optionally substituted with 1 or 2 groups independently selected from C 1-4 alkyl and C 1-4 alkoxy, wherein the C 1-4 alkyl group is optionally substituted by OH.
  • R 7 is H, piperidin-1-yl, or pyridin-3-yl, wherein the piperidin-1-yl and piperidin-3-yl groups are each optionally substituted with 1 or 2 groups independently selected from C 1-4 alkyl and C 1-4 alkoxy, wherein the C 1-4 alkyl group is optionally substituted by OH.
  • R 7 , R 8 , and R 9 are each H.
  • the compound is a compound of Formula X, XI, or XII: or a pharmaceutically acceptable salt thereof, wherein:
  • the compound is a compound of Formula X, XI, or XII: or a pharmaceutically acceptable salt thereof, wherein:
  • the compound is a compound of Formula X, XI, or XII: or a pharmaceutically acceptable salt thereof, wherein:
  • the compound is a compound of Formula X, XI, or XII: or a pharmaceutically acceptable salt thereof, wherein:
  • the compound is a compound of Formula X, XI, or XII: or a pharmaceutically acceptable salt thereof, wherein:
  • the compound is a compound of Formula X, XI, or XII: or a pharmaceutically acceptable salt thereof, wherein:
  • the compound is a compound of Formula X, XI, or XII: or a pharmaceutically acceptable salt thereof, wherein:
  • the compound of the invention is a compound of Formula (II): or a pharmaceutically acceptable salt thereof.
  • the compound is a compound of Formula (III): or a pharmaceutically acceptable salt thereof.
  • the compound is a compound of Formula (IV): or a pharmaceutically acceptable salt or stereoisomer thereof.
  • the compound is a compound of Formula (V): or a pharmaceutically acceptable salt thereof.
  • the compound is a compound of Formula (VI): or a pharmaceutically acceptable salt thereof.
  • the compound is a compound of Formula (VII): or a pharmaceutically acceptable salt thereof.
  • the compound is a compound of Formula (VIII): or a pharmaceutically acceptable salt thereof.
  • the compound is a compound of Formula (IX): or a pharmaceutically acceptable salt thereof.
  • the compound is a compound of Formula (X): or a pharmaceutically acceptable salt thereof.
  • the compound is a compound of Formula (X), or a pharmaceutically acceptable salt thereof, wherein R 4a is H, C 1-6 alkyl, C 2-6 alkynyl, benzyl, cyanomethyl, 2-methoxyethyl, 2-hydroxyethyl, cyclobutyl, cyclopentyl, 2-amino-2-oxoethyl, N,N-dimethyl-2-amino-2-oxoethyl, (1-methyl-1H-pyrazol-3-yl)methyl, 2-morpholinoethyl, 2-morpholino-2-oxoethyl or 2-aminoethyl.
  • R 4a is H, C 1-6 alkyl, C 2-6 alkynyl, benzyl, cyanomethyl, 2-methoxyethyl, 2-hydroxyethyl, cyclobutyl, cyclopentyl, 2-amino-2-oxoethyl, N,N-dimethyl-2
  • the compound is a compound of Formula (X), or a pharmaceutically acceptable salt thereof, wherein R 2 is ethoxy or phenyl optionally substituted with halo.
  • the compound is a compound of Formula (X), or a pharmaceutically acceptable salt thereof, wherein R 1 is methyl.
  • the compound is a compound of Formula (X), or a pharmaceutically acceptable salt thereof, wherein R 5 is H, Br or methyl.
  • the compound is a compound of Formula (X), or a pharmaceutically acceptable salt thereof, wherein R 6 is methyl, CN or Cl.
  • the compound is a compound of Formula (X), or a pharmaceutically acceptable salt thereof, wherein R 7 , R 8 and R 9 are each H.
  • the compound is a compound of Formula (XI): or a pharmaceutically acceptable salt thereof.
  • the compound is a compound of Formula (XI), or a pharmaceutically acceptable salt thereof, wherein R 4a is H, C 1-6 alkyl, C 2-6 alkynyl, benzyl, cyanomethyl, 2-methoxyethyl, 2-hydroxyethyl, cyclobutyl, cyclopentyl, 2-amino-2-oxoethyl, N,N-dimethyl-2-amino-2-oxoethyl, (1-methyl-1H-pyrazol-3-yl)methyl, 2-morpholinoethyl, 2-morpholino-2-oxoethyl or 2-aminoethyl.
  • R 4a is H, C 1-6 alkyl, C 2-6 alkynyl, benzyl, cyanomethyl, 2-methoxyethyl, 2-hydroxyethyl, cyclobutyl, cyclopentyl, 2-amino-2-oxoethyl, N,N-dimethyl
  • the compound is a compound of Formula (XI), or a pharmaceutically acceptable salt thereof, wherein R 2 is ethoxy, 1,1-dioxidothiomorpholino optionally substituted with 1 or 2 independently selected C 1-4 alkyl substituents, or phenyl optionally substituted with halo.
  • the compound is a compound of Formula (XI), or a pharmaceutically acceptable salt thereof, wherein R 2 is ethoxy or phenyl optionally substituted with halo.
  • the compound is a compound of Formula (XI), or a pharmaceutically acceptable salt thereof, wherein R 1 is methyl.
  • the compound is a compound of Formula (XI), or a pharmaceutically acceptable salt thereof, wherein R 5 is H, Br or methyl.
  • the compound is a compound of Formula (XI), or a pharmaceutically acceptable salt thereof, wherein R 6 is methyl, CN or Cl.
  • the compound is a compound of Formula (XI), or a pharmaceutically acceptable salt thereof, wherein R 7 is H, piperidin-1-yl, or pyridin-3-yl optionally substituted by C 1-4 alkyl or C 1-4 alkoxy, wherein the C 1-4 alkyl group is substituted by OH.
  • the compound is a compound of Formula (XI), or a pharmaceutically acceptable salt thereof, wherein R 7 , R 8 and R 9 are each H.
  • the compound is a compound of Formula (XII): or a pharmaceutically acceptable salt thereof.
  • the compound is 2-amino-N-((S)-1-(8-chloro-5-((R)-2-methyl-1,1-dioxidothiomorpholino)imidazo[1,5-a]pyridin-6-yl)ethyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide, or a pharmaceutically acceptable salt thereof.
  • the compound is 2-amino-N-((S)-1-(8-chloro-5-((S)-2-methyl-1,1-dioxidothiomorpholino)imidazo[1,5-a]pyridin-6-yl)ethyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide, or a pharmaceutically acceptable salt thereof.
  • n-membered where n is an integer typically describes the number of ring-forming atoms in a moiety where the number of ring-forming atoms is n.
  • piperidinyl is an example of a 6-membered heterocycloalkyl ring
  • pyrazolyl is an example of a 5-membered heteroaryl ring
  • pyridyl is an example of a 6-membered heteroaryl ring
  • 1,2,3,4-tetrahydro-naphthalene is an example of a 10-membered cycloalkyl group.
  • the phrase "optionally substituted” means unsubstituted or substituted.
  • the substituents are independently selected, and substitution may be at any chemically accessible position.
  • substituted means that a hydrogen atom is removed and replaced by a substituent.
  • a single divalent substituent, e.g., oxo, can replace two hydrogen atoms. It is to be understood that substitution at a given atom is limited by valency.
  • C n-m indicates a range which includes the endpoints, wherein n and m are integers and indicate the number of carbons. Examples include C 1-4 , C 1-6 , and the like.
  • C n-m alkyl refers to a saturated hydrocarbon group that may be straight-chain or branched, having n to m carbons.
  • alkyl moieties include, but are not limited to, chemical groups such as methyl, ethyl, n -propyl, isopropyl, n -butyl, tert -butyl, isobutyl, sec -butyl; higher homologs such as 2-methyl-1-butyl, n -pentyl, 3-pentyl, n -hexyl, 1,2,2-trimethylpropyl, and the like.
  • the alkyl group contains from 1 to 6 carbon atoms, from 1 to 4 carbon atoms, from 1 to 3 carbon atoms, or 1 to 2 carbon atoms.
  • C n-m alkenyl refers to an alkyl group having one or more double carbon-carbon bonds and having n to m carbons.
  • Example alkenyl groups include, but are not limited to, ethenyl, n -propenyl, isopropenyl, n -butenyl, sec -butenyl, and the like.
  • the alkenyl moiety contains 2 to 6, 2 to 4, or 2 to 3 carbon atoms.
  • C n-m alkynyl refers to an alkyl group having one or more triple carbon-carbon bonds and having n to m carbons.
  • Example alkynyl groups include, but are not limited to, ethynyl, propyn-1-yl, propyn-2-yl, and the like.
  • the alkynyl moiety contains 2 to 6, 2 to 4, or 2 to 3 carbon atoms.
  • C n-m alkylene refers to a divalent alkyl linking group having n to m carbons.
  • alkylene groups include, but are not limited to, ethan-1,1-diyl, ethan-1,2-diyl, propan-1,1,-diyl, propan-1,3-diyl, propan-1,2-diyl, butan-1,4-diyl, butan-1,3-diyl, butan-1,2-diyl, 2-methyl-propan-1,3-diyl, and the like.
  • the alkylene moiety contains 2 to 6, 2 to 4, 2 to 3, 1 to 6, 1 to 4, or 1 to 2 carbon atoms.
  • C n-m alkoxy refers to a group of formula -O-alkyl, wherein the alkyl group has n to m carbons.
  • Example alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy (e.g., n -propoxy and isopropoxy), butoxy (e.g., n -butoxy and tert -butoxy), and the like.
  • the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.
  • C n-m alkylamino refers to a group of formula -NH(alkyl), wherein the alkyl group has n to m carbon atoms. In some embodiments, the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.
  • alkylamino groups include, but are not limited to, N-methylamino, N-ethylamino, N-propylamino (e.g., N-( n -propyl)amino and N-isopropylamino), N-butylamino (e.g., N-( n -butyl)amino and N-( tert -butyl)amino), and the like.
  • amino refers to a group of formula -NH 2 .
  • aryl employed alone or in combination with other terms, refers to an aromatic hydrocarbon group, which may be monocyclic or polycyclic (e.g., having 2, 3 or 4 fused rings).
  • C n-m aryl refers to an aryl group having from n to m ring carbon atoms.
  • Aryl groups include, e.g., phenyl, naphthyl, anthracenyl, phenanthrenyl, indanyl, indenyl, and the like. In some embodiments, aryl groups have from 6 to 10 carbon atoms. In some embodiments, the aryl group is phenyl or naphthyl.
  • di(C n-m -alkyl)amino refers to a group of formula -N(alkyl) 2 , wherein the two alkyl groups each has, independently, n to m carbon atoms. In some embodiments, each alkyl group independently has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.
  • halo refers to F, Cl, Br, or I. In some embodiments, a halo is F, Cl, or Br.
  • C n-m haloalkoxy refers to a group of formula -O-haloalkyl having n to m carbon atoms.
  • An example haloalkoxy group is OCF 3 .
  • the haloalkoxy group is fluorinated only.
  • the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.
  • C n-m haloalkyl refers to an alkyl group having from one halogen atom to 2s+1 halogen atoms which may be the same or different, where "s" is the number of carbon atoms in the alkyl group, wherein the alkyl group has n to m carbon atoms.
  • the haloalkyl group is fluorinated only.
  • the alkyl group has 1 to 6, 1 to 4, or 1 to 3 carbon atoms.
  • cycloalkyl refers to non-aromatic cyclic hydrocarbons including cyclized alkyl and/or alkenyl groups.
  • Cycloalkyl groups can include mono- or polycyclic (e.g., having 2, 3 or 4 fused rings) groups and spirocycles. Cycloalkyl groups can have 3, 4, 5, 6, 7, 8, 9, or 10 ring-forming carbons (C 3-10 ).
  • the cycloalkyl is a C 3-10 monocyclic or bicyclic cyclocalkyl.
  • the cycloalkyl is a C 3-7 monocyclic cyclocalkyl.
  • Example cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptatrienyl, norbornyl, norpinyl, norcarnyl, and the like.
  • cycloalkyl is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.
  • heteroaryl refers to a monocyclic or polycyclic aromatic heterocycle having at least one heteroatom ring member selected from sulfur, oxygen, and nitrogen.
  • the heteroaryl ring has 1, 2, 3, or 4 heteroatom ring members independently selected from nitrogen, sulfur and oxygen.
  • any ring-forming N in a heteroaryl moiety can be an N-oxide.
  • the heteroaryl is a 5-10 membered monocyclic or bicyclic heteroaryl having 1, 2, 3 or 4 heteroatom ring members independently selected from nitrogen, sulfur and oxygen.
  • the heteroaryl is a 5-6 monocyclic heteroaryl having 1 or 2 heteroatom ring members independently selected from nitrogen, sulfur and oxygen.
  • the heteroaryl is a five-membered or six-membereted heteroaryl ring.
  • a five-membered heteroaryl ring is a heteroaryl with a ring having five ring atoms wherein one or more (e.g., 1, 2, or 3) ring atoms are independently selected from N, O, and S.
  • Exemplary five-membered ring heteroaryls are thienyl, furyl, pyrrolyl, imidazolyl, thiazolyl, oxazolyl, pyrazolyl, isothiazolyl, isoxazolyl, 1,2,3-triazolyl, tetrazolyl, 1,2,3-thiadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-triazolyl, 1,2,4-thiadiazolyl, 1,2,4-oxadiazolyl, 1,3,4-triazolyl, 1,3,4-thiadiazolyl, and 1,3,4-oxadiazolyl.
  • a six-membered heteroaryl ring is a heteroaryl with a ring having six ring atoms wherein one or more (e.g., 1, 2, or 3) ring atoms are independently selected from N, O, and S.
  • Exemplary six-membered ring heteroaryls are pyridyl, pyrazinyl, pyrimidinyl, triazinyl and pyridazinyl.
  • heterocycloalkyl refers to non-aromatic monocyclic or polycyclic heterocycles having one or more ring-forming heteroatoms selected from O, N, or S. Included in heterocycloalkyl are monocyclic 4-, 5-, 6-, 7-, 8-, 9- or 10-membered heterocycloalkyl groups. Heterocycloalkyl groups can also include spirocycles.
  • Example heterocycloalkyl groups include pyrrolidin-2-one, 1,3-isoxazolidin-2-one, pyranyl, tetrahydropuran, oxetanyl, azetidinyl, morpholino, thiomorpholino, 1,1-dioxidothiomorpholin-4-yl, 1,2,3,6-tetrahydropyridin-4-yl, 1,1-dioxido-1,4-thiazepan-4-yl, 1,1-dioxide-1,2,5-thiadiazepan-5-yl, piperazinyl, tetrahydrofuranyl, tetrahydrothienyl, piperidinyl, pyrrolidinyl, isoxazolidinyl, isothiazolidinyl, pyrazolidinyl, oxazolidinyl, thiazolidinyl, imidazolidinyl, azepanyl, benzazapene,
  • Ring-forming carbon atoms and heteroatoms of a heterocycloalkyl group can be optionally substituted by oxo or sulfido (e.g., C(O), S(O), C(S), or S(O) 2 , etc.).
  • the heterocycloalkyl group can be attached through a ring-forming carbon atom or a ring-forming heteroatom.
  • the heterocycloalkyl group contains 0 to 3 double bonds. In some embodiments, the heterocycloalkyl group contains 0 to 2 double bonds.
  • heterocycloalkyl moieties that have one or more aromatic rings fused ( i.e ., having a bond in common with) to the cycloalkyl ring, for example, benzo or thienyl derivatives of piperidine, morpholine, azepine, etc.
  • a heterocycloalkyl group containing a fused aromatic ring can be attached through any ring-forming atom including a ring-forming atom of the fused aromatic ring.
  • the heterocycloalkyl is a monocyclic 4-6 membered heterocycloalkyl having 1 or 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur and having one or more oxidized ring members.
  • the heterocycloalkyl is a monocyclic or bicyclic 4-10 membered heterocycloalkyl having 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur and having one or more oxidized ring members. In some embodiments, the heterocycloalkyl is a monocyclic or bicyclic 4-10 membered heterocycloalkyl having 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur and having one or more ring members selected from C(O), S(O), C(S), S(O) 2 , and S(NH)(O).
  • the heterocycloalkyl is a monocyclic or bicyclic 4-10 membered heterocycloalkyl having 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur and having one or more ring members selected from S(O) 2 and S(NH)(O).
  • the definitions or embodiments refer to specific rings (e.g., an azetidine ring, a pyridine ring, etc.). Unless otherwise indicated, these rings can be attached to any ring member provided that the valency of the atom is not exceeded. For example, an azetidine ring may be attached at any position of the ring, whereas a pyridin-3-yl ring is attached at the 3-position.
  • the compounds described herein can be asymmetric ( e . g ., having one or more stereocenters). All stereoisomers, such as enantiomers and diastereomers, are intended unless otherwise indicated.
  • Cis and trans geometric isomers of the compounds of the present invention are described and may be isolated as a mixture of isomers or as separated isomeric forms.
  • the compound has the (R)- configuration.
  • the compound has the (S)- configuration.
  • Formulas (I)-(XII) herein include stereoisomers of the compounds.
  • the carbon atom to which R 1 is attached is in the ( R )-configuration.
  • the carbon atom to which R 1 is attached is in the (S) -configuration.
  • An example method includes fractional recrystallizaion using a chiral resolving acid which is an optically active, salt-forming organic acid.
  • Suitable resolving agents for fractional recrystallization methods are, for example, optically active acids, such as the D and L forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid or the various optically active camphorsulfonic acids such as ⁇ -camphorsulfonic acid.
  • Other resolving agents suitable for fractional crystallization methods include stereoisomerically pure forms of ⁇ -methylbenzylamine (e.g.
  • Resolution of racemic mixtures can also be carried out by elution on a column packed with an optically active resolving agent (e . g ., dinitrobenzoylphenylglycine).
  • an optically active resolving agent e . g ., dinitrobenzoylphenylglycine.
  • Suitable elution solvent composition can be determined by one skilled in the art.
  • Tautomeric forms result from the swapping of a single bond with an adjacent double bond together with the concomitant migration of a proton.
  • Tautomeric forms include prototropic tautomers which are isomeric protonation states having the same empirical formula and total charge.
  • Example prototropic tautomers include ketone - enol pairs, amide - imidic acid pairs, lactam - lactim pairs, enamine - imine pairs, and annular forms where a proton can occupy two or more positions of a heterocyclic system, for example, 1H- and 3H-imidazole, 1H-, 2H- and 4H- 1,2,4-triazole, 1H- and 2H- isoindole, and 1H- and 2H-pyrazole.
  • Tautomeric forms can be in equilibrium or sterically locked into one form by appropriate substitution.
  • All compounds, and pharmaceutically acceptable salts thereof, can be found together with other substances such as water and solvents (e.g. hydrates and solvates) or can be isolated.
  • preparation of compounds can involve the addition of acids or bases to affect, for example, catalysis of a desired reaction or formation of salt forms such as acid addition salts.
  • Example acids can be inorganic or organic acids and include, but are not limited to, strong and weak acids.
  • Some example acids include hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, p -toluenesulfonic acid, 4-nitrobenzoic acid, methanesulfonic acid, benzenesulfonic acid, trifluoroacetic acid, and nitric acid.
  • Some weak acids include, but are not limited to acetic acid, propionic acid, butanoic acid, benzoic acid, tartaric acid, pentanoic acid, hexanoic acid, heptanoic acid, octanoic acid, nonanoic acid, and decanoic acid.
  • Example bases include lithium hydroxide, sodium hydroxide, potassium hydroxide, lithium carbonate, sodium carbonate, potassium carbonate, and sodium bicarbonate.
  • Some example strong bases include, but are not limited to, hydroxide, alkoxides, metal amides, metal hydrides, metal dialkylamides and arylamines, wherein; alkoxides include lithium, sodium and potassium salts of methyl, ethyl and t-butyl oxides; metal amides include sodium amide, potassium amide and lithium amide; metal hydrides include sodium hydride, potassium hydride and lithium hydride; and metal dialkylamides include lithium, sodium, and potassium salts of methyl, ethyl, n-propyl, iso -propyl, n-butyl, tert -butyl, trimethylsilyl and cyclohexyl substituted amides.
  • the compounds provided herein, or salts thereof are substantially isolated.
  • substantially isolated is meant that the compound is at least partially or substantially separated from the environment in which it was formed or detected.
  • Partial separation can include, for example, a composition enriched in the compounds provided herein.
  • Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compounds provided herein, or salt thereof. Methods for isolating compounds and their salts are routine in the art.
  • Compounds of the invention can also include all isotopes of atoms occurring in the intermediates or final compounds.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include tritium and deuterium.
  • One or more constituent atoms of the compounds of the invention can be replaced or substituted with isotopes of the atoms in natural or non-natural abundance.
  • the compound includes at least one deuterium atom.
  • one or more hydrogen atoms in a compound of the present disclosure can be replaced or substituted by deuterium.
  • the compound includes two or more deuterium atoms.
  • the compound includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 deuterium atoms. Synthetic methods for including isotopes into organic compounds are known in the art.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • the present application also includes pharmaceutically acceptable salts of the compounds described herein.
  • the present invention also includes pharmaceutically acceptable salts of the compounds described herein.
  • pharmaceutically acceptable salts refers to derivatives of the disclosed compounds wherein the parent compound is modified by converting an existing acid or base moiety to its salt form.
  • pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts of the present invention include the conventional non-toxic salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods.
  • such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred.
  • non-aqueous media like ether, ethyl acetate, alcohols (e.g., methanol, ethanol, iso-propanol, or butanol) or acetonitrile (ACN) are preferred.
  • ACN acetonitrile
  • the compounds provided herein can be prepared using known organic synthesis techniques and can be synthesized according to any of numerous possible synthetic routes.
  • optionally chosen protecting groups can be removed under conditions suitable for their removal, that are also compatible with the functionality present in the resulting compounds of Formula I and compounds of the invention (compounds of Formula I wherein Y 1 is N, Y 2 is N, Z 1 is C, Z 2 is N).
  • Amines 1-1 can be prepared from ketones 2-1 by various methods, as illustrated in Scheme 2 .
  • one method ( A ) involves imine formation with a source of ammonia (e . g . solution of ammonia in an alcohol, or an ammonium salt such as ammonium acetate), which may be facilitated by the introduction of a Lewis acid (e . g . titanium(IV) ethoxide or titanium tetraisopropoxide).
  • the imine can be reduced using a reducing agent ( e . g . NaBH4 or NaCNBH3), to furnish amines 1-1 .
  • the method of Ellman ( B ) can be used to furnish amine 1-1 in enantiomerically enriched form.
  • ketones 2-1 with a tert- butanesulfinamide (chiral, if desired) in the presence of Lewis acid (e . g ., titanium tetraethoxide), followed by reduction of the tert -butanesulfinyl ketimine ( e . g . using L-Selectride ® ), and removal of the tert -butyl sulfinyl group with acid ( e.g. , 4 N HCl in dioxane).
  • Lewis acid e. g ., titanium tetraethoxide
  • tert -butanesulfinyl ketimine e . g . using L-Selectride ®
  • removal of the tert -butyl sulfinyl group with acid e.g. , 4 N HCl in dioxane.
  • a sequence of transformations ( C ) can be used to furnish amine 1-1 , which include reduction of the ketone to the alcohol (e . g . using NaBH 4 ), conversion of the resulting alcohol to a leaving group ( e . g ., reacting with MsCl in the presence of base, such as triethylamine), displacement of the leaving group with sodium azide, followed by reduction of the azide to an amine ( e . g . , via hydrogenation or Staudinger reduction).
  • base such as triethylamine
  • ketones 2-1 can be prepared from esters 3-1 via sequential hydrolysis (e.g. treatment with a hydroxide base, such as NaOH), Weinreb amide formation ( e.g. by coupling the acid with N,O -dimethylhydroxylamine using a coupling agent such as EDCI and HOBt in the presence of a tertiary amine base, such as triethylamine or diisopropylethylamine), and treatment of the Weinreb amide with a Grignard reagent, R 1 -MgX. to afford ketone 2-1 .
  • a hydroxide base such as NaOH
  • Weinreb amide formation e.g. by coupling the acid with N,O -dimethylhydroxylamine using a coupling agent such as EDCI and HOBt in the presence of a tertiary amine base, such as triethylamine or diisopropylethylamine
  • a Grignard reagent R 1
  • the substituent R 2 can be introduced by a coupling of 4-2 with R 2 -M, where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu)4 or Zn.
  • M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu)4 or Zn.
  • Suzuki conditions e.g. , in the presence of a palladium catalyst, such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)
  • a base e.g. , a bicarbonate or carbonate base, or CsF
  • Stille conditions e.
  • a palladium(0) catalyst such as tetrakis(triphenylphosphine)palladium(0)
  • standard Negishi conditions e . g ., in the presence of a palladium catalyst, such as tetrakis(triphenylphosphine)palladium(0) or [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium (II)
  • a palladium(0) catalyst such as tetrakis(triphenylphosphine)palladium(0) or [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium (II)
  • the nitro group of intermediate 4-3 can be converted to the primary amine by reduction ( e . g . hydrogenation over a Pt or Pd catalyst, Fe/HCl, or LiAlH 4 ).
  • Intermediate 4-4 can then be converted to the indazole via diazotization of the amine and cyclization employing an alkyl nitrite (e . g ., amyl nitrite) in warm AcOH.
  • an alkyl nitrite e . g ., amyl nitrite
  • the halogen present in 4-5 can serve as a handle for installation of substituent R 6 , via coupling with R 6 -M, where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu) 4 or Zn, under standard Suzuki conditions ( e .
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)) or standard Stille conditions ( e.g. , in the presence of a palladium(0) catalyst, such as tetrakis(triphenylphosphine)palladium(0)) or standard Negishi conditions ( e .
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium (II)
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium (II)
  • the indazole nitrogens of 4-6 can be alkylated either by treatment with a base and an electrophile, such as an alkyl halide (e.g. K 2 CO 3 and MeI), or via Mitsunobu conditions (PPh 3 , DEAD) employing a suitable alcohol, to give 4-7 as a mixture of N1 and N2 substituted indazoles.
  • an alkyl halide e.g. K 2 CO 3 and MeI
  • Mitsunobu conditions PPh 3
  • the ketone in 4-7 can be converted to an amine by various methods as shown in Scheme 2 to furnish amine 4-8 .
  • Amine 4-8 can be coupled with an optionally protected carboxylic acid such as 1-2 (from Scheme 1 ) by various methods as shown in Scheme 1 .
  • any chosen protecting groups can be removed under conditions suitable for their removal, that are also compatible with the functionality present in the resulting compounds of the Formula I and compounds of the invention (compounds of Formula I wherein Y 1 is N, Y 2 is N, Z 1 is C, Z 2 is N). It will be recognized by one skilled in the art that the order of steps in Scheme 4 can be changed in consideration of compatibility of functional groups present in the intermediates.
  • Alkylation of the hydroxyl group in 4-1 employing a base and an electrophile such as an alkyl halide (e.g. K 2 CO 3 and MeI) provides alkoxy intermediates 5-1 , which can be converted to compounds of Formula I and compounds of the invention (compounds of Formula I wherein Y 1 is N, Y 2 is N, Z 1 is C, Z 2 is N) employing the synthetic methodologies described in Schemes 1 and 2. It will be recognized by one skilled in the art that the order of steps in Scheme 5 can be changed in consideration of the compatibility of functional groups present in the intermediates.
  • the ketone 4-6 can be converted to an amine 6-1 by various methods as outlined in Scheme 2 .
  • Amine 6-1 can be coupled with an optionally protected carboxylic acid such as 1-2 (from Scheme 1 ) by various methods ( e .
  • a coupling reagent such as N,N,N' , N' -tetramethyl- O -(7-azabenzotriazol-1-yl)uronium hexafluorophosphate in the presence of base, such as diisopropylethylamine).
  • the indazole nitrogens of 6-2 can be alkylated either by treatment with a base and an electrophile such as an alkyl halide ( e . g . K 2 CO 3 and MeI), or via Mitsunobu conditions (PPh 3 , DEAD) employing a suitable alcohol, to give 6-3 as a mixture of N1 and N2 substituted indazoles.
  • Any chosen protecting groups can then be removed under conditions suitable for their removal that are also compatible with the functionality present in the resulting compounds of the invention. It will be recognized by one skilled in the art that the order of steps in Scheme 6 can be changed in consideration of compatibility of functional groups present in the intermediates.
  • Halogenation of heterocycle 4-6 with reagents such as I 2 , Br 2 , N -bromosuccinimide, or N -iodosuccinimide can furnish intermediate 7-1 , which can serve as a substrate for introduction of substituent R 5 , via coupling with R 5 -M, where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu)4 or Zn, under standard Suzuki conditions ( e . g ., in the presence of a palladium catalyst, such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base (e.g.
  • Amine 7-3 can be coupled with an optionally protected carboxylic acid such as 1-2 (from Scheme 1 ) by various methods ( e . g . , treatment with a coupling reagent such as N,N,N' , N' -tetramethyl- O -(7-azabenzotriazol-1-yl)uronium hexafluorophosphate in the presence of base, such as diisopropylethylamine) to provide 7-4 .
  • the indazole nitrogens in 7-4 can optionally be alkylated either by treatment with a base and an electrophile, such as an alkyl halide ( e . g .
  • the substituent R 2 can subsequently be introduced by coupling with R 2 -M, where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu) 4 or Zn, under standard Suzuki conditions ( e . g ., in the presence of a palladium catalyst, such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)) or standard Stille conditions ( e.g.
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)
  • base e.g. , a bicarbonate
  • halogen present in 8-7 can serve as a handle for installation of substituent R 5 , via coupling with R 5 -M, where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu)4 or Zn, under standard Suzuki conditions ( e . g ., in the presence of a palladium catalyst, such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g.
  • ketone 8-9 from ester 8-8 can be achieved by several methods such as those described in Scheme 3 .
  • Halo-containing starting materials (9-1) can be esterified by various methods ( e . g ., conversion of the carboxylic acid to the acid chloride by reaction with COCl 2 and DMF in a suitable solvent such as DCM, followed by reaction of the acid chloride with a suitable alcohol, such as EtOH).
  • the ester intermediate can then be treated with an oxidizing reagent (e.g. , a peroxide reagent such as the combination of H 2 O 2 /TFA) to form the heterocyclic N- oxide, and subsequently be converted to the nitrile derivative 9-2 ( e.g. , by heating with trimethylsilylcyanide and base).
  • an oxidizing reagent e.g. , a peroxide reagent such as the combination of H 2 O 2 /TFA
  • the substituent R 2 can be introduced by a coupling of 9-2 with R 2 -M, where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu) 4 or Zn, under standard Suzuki conditions ( e .
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)) or standard Stille conditions ( e.g. , in the presence of a palladium(0) catalyst, such as tetrakis(triphenylphosphine)palladium(0)) or standard Negishi conditions ( e .
  • nitrile of intermediate 9-3 can be converted to an aminomethyl group by reduction ( e . g . using H 2 and a catalyst, such as Raney ® Ni or Pd on carbon).
  • the aminomethyl group can be acylated ( e . g . by reacting with R 3 -CO-LG, wherein CO-LG is a suitable activated carbonyl group ( e.g.
  • the acylated intermediate can be cyclized to form the bicyclic intermediate 9-4 under cyclo-dehydrating conditions ( e.g. by heating in POCl 3 or by treatment with P 2 O 5 , SOCl 2 or with acid). Formation of ketone 9-5 from ester 9-4 can proceed as illustrated in Scheme 3 . If desired, the halogen present in 9-5 (i.e.
  • group X can serve as a handle for installation of substituent R 6 , via coupling with R 6 -M, where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu) 4 or Zn, under standard Suzuki conditions ( e . g ., in the presence of a palladium catalyst, such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)) or standard Stille conditions ( e .
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)
  • base e.g.
  • a palladium(0) catalyst such as tetrakis(triphenylphosphine)palladium(0)
  • standard Negishi conditions e . g ., in the presence of a palladium catalyst, such as tetrakis(triphenylphosphine)palladium(0) or [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium (II)
  • a palladium(0) catalyst such as tetrakis(triphenylphosphine)palladium(0) or [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium (II)
  • Ketone 9-8 can be treated with one of the conditions of Scheme 2 to provide an amine that can be converted to compounds of the invention 9-9 according to Scheme 1 . It will be recognized by one skilled in the art that the order of steps in Scheme 9 can be changed in consideration of compatibility of functional groups present in the intermediates.
  • the amide intermediate 9b-1 can then be treated with an oxidizing reagent (e . g ., a peroxide reagent such as the combination of H 2 O 2 /TFA) to form the heterocyclic N -oxide, and subsequently be converted to the nitrile derivative 9b-1 ( e.g. , by heating with trimethylsilylcyanide and base).
  • an oxidizing reagent e. g ., a peroxide reagent such as the combination of H 2 O 2 /TFA
  • the nitrile of intermediate 9b-1 can be converted to an aminomethyl group by reduction ( e . g . Raney ® Ni in formic acid).
  • the acylated intermediate can be cyclized to form the bicyclic intermediate 9b-2 under cyclo-dehydrating conditions ( e . g .
  • the Weinreb amide of intermediate 9b-2 can undergo reaction with a Grignard reagent R 1 MgX to afford ketone 9b-3 .
  • the substituent R 2 can be introduced by a coupling of 9b-3 with R 2 -M, where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu) 4 or Zn, under standard Suzuki conditions ( e .
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)) or standard Stille conditions ( e . g ., in the presence of a palladium(0) catalyst, such as tetrakis(triphenylphosphine)palladium(0)) or standard Negishi conditions ( e .
  • the imidazole ring can be annealed to provide intermediate 9c-2 , which can be elaborated to ketone 9c-3 , which can be functionalized to afford 9c-4, 9c-5 and 9c-6 , and finally elaborated to compounds of Formula I and compounds of the invention (compounds of Formula I wherein Y 1 is N, Y 2 is N, Z 1 is C, Z 2 is N) using methods previously outlined in Scheme 9 .
  • intermediate 9c-3 can be further functionalized and elaborated to compounds of Formula I and compounds of the invention (compounds of Formula I wherein Y 1 is N, Y 2 is N, Z 1 is C, Z 2 is N) using methods already described for Scheme 9 and Scheme 9c .
  • acetonitrile furnishes adduct 9e-1 , which is very flexible for further transformations.
  • Intermediate Weinreb amide 9e-1 can undergo reaction with Grignard reagents R 1 MgX to afford ketones of type 9e-2 that can be treated according to the methods of Scheme 2 to afford amines 9e-5 which, in turn, can be converted to compounds of Formula I and compounds of the invention (compounds of Formula I wherein Y 1 is N, Y 2 is N, Z 1 is C, Z 2 is N) as outlined in Scheme 1 .
  • Weinreb amide 9e-1 can be reduced by a suitable reducing agent ( e . g .
  • aldehyde 9e-3 can either be converted to amine 9e-4 by one of the methods of Scheme 2 , or converted to amine 9e-5 by condensation with tert -butylsulfinamide (chiral, if desired) in the presence of lewis acid (e . g . titanium isopropoxide), followed by reaction of the tert -butanesulfinyl aldimine with a Grignard reagent R 1 MgX, and removal of the tert -butyl sulfinyl group with acid (e.g. 4N HCl in dioxane).
  • lewis acid e . g . titanium isopropoxide
  • Amines 9e-4 and 9e-5 serve as useful intermediates for further functionalization and elaboration to compounds of Formula I and compounds of the invention (compounds of Formula I wherein Y 1 is N, Y 2 is N, Z 1 is C, Z 2 is N) according to methods outlined in Scheme 9 . It will be recognized by one skilled in the art that substituents at R 5 and R 6 can be introduced on many of the intermediates at various stages in Scheme 9d and Scheme 9e , according to the compatibility of functional groups with the subsequent steps to be performed.
  • Substituent R 2 can be introduced, for example, via cross-coupling with R 2 -M, where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu) 4 or Zn, under standard Suzuki conditions (e . g ., in the presence of a palladium catalyst, such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)) or standard Stille conditions (e.g.
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)
  • base e.g. ,
  • R 2 can be installed via nucleophilic aromatic substitution of the halogen of 10-2 with amines, for example, to provide 10-3 where R 2 is an amine. Halogenation of 10-3 using a halogenating reagent (e.g.
  • a suitable base e. g ., Na 2 CO 3
  • the intermediate 10-5 may be in the form of the acetal or ketal, in which case reaction of such intermediate with 10-4 may include acid rather than base ( e.g. , p- toluenesulfonic acid).
  • the halogen in 10-7 can be used as a handle for the introduction of R 6 via coupling with M-R 6 , where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu) 4 or Zn, under standard Suzuki conditions ( e . g ., in the presence of a palladium catalyst, such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base (e.g.
  • Ketone 10-8 can be treated with one of the conditions of Scheme 2 to provide an amine that can be converted to compounds of the invention 10-9 according to Scheme 1 . It will be recognized by one skilled in the art that the order of steps in Scheme 10 can be changed in consideration of compatibility of functional groups present in the intermediates.
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)) or standard Stille conditions ( e.g. , in the presence of a palladium(0) catalyst, such as tetrakis(triphenylphosphine)palladium(0)) or standard Negishi conditions ( e .
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium (II)
  • a suitable oxidant e.g., m -CPBA
  • a suitable reagent e.g., POX 3 , RCOX, RSO 2 X
  • Nucleophilic aromatic substitution of the halide (i.e. , group X) in intermediate 11-3 with hydrazine can afford intermediate 11-4 .
  • a coupling agent e.g. , BOP, HATU, or EDCI/HOBt
  • a base e.g. , diisopropylethylamine
  • Direct conversion of halide 11-3 to acylhydrazide intermediates 11-6 is also possible by displacement of the halide of 11-3 directly with an acyl hydrazide (e . g ., R 3 (CO)NHNH 2 ).
  • a palladium-catalyzed cross coupling of an acyl hydrazide ( e.g. , R 3 (CO)NHNH 2 ) with heteroaryl halide 11-3 e.g. , as described in Org. Lett. 2010, 12(4), p.792-795 ; using Pd 2 (dba) 3 , Josiphos, NaHCO 3 in DMF at elevated temperature
  • the intermediate 11-6 can be cyclized to form bicyclic intermediate 11-7 under cyclo-dehydrating conditions (e . g . by heating in POCl 3 or by treatment with P 2 O 5 , SOCl 2 or with acid).
  • cyclo-dehydrating conditions e . g . by heating in POCl 3 or by treatment with P 2 O 5 , SOCl 2 or with acid.
  • hydrazine containing intermediate 11-4 can be heated with orthoesters (e . g ., trimethylorthoformate, triethylorthoacetate, or R 3 C(OR) 3 ), often under acid catalysis ( e . g ., p-toluenesulfonic acid) to furnish 11-7 directly.
  • orthoesters e . g ., trimethylorthoformate, triethylorthoacetate, or R 3 C(OR) 3
  • acid catalysis e . g ., p-tol
  • R 6 can be introduced into the compound 11-7 via cross-coupling with R 6 -M, where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu) 4 or Zn, under standard Suzuki conditions ( e . g ., in the presence of a palladium catalyst, such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)) or standard Stille conditions ( e .
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)
  • base e.g. , a bicarbon
  • Halogenation e . g ., NCS
  • intermediates 12-5 can undergo cross coupling with tributylstannyl vinyl ethers under standard Stille conditions ( e . g ., in the presence of a palladium(0) catalyst, such as tetrakis(triphenylphosphine)palladium(0), in addition to a base such as CsF) to furnish intermediates 12-7 after hydrolysis.
  • a palladium(0) catalyst such as tetrakis(triphenylphosphine)palladium(0)
  • a base such as CsF
  • Nucleophilic aromatic substitution with ammonia, followed by halogenation then affords 12-9 .
  • Sandmeyer reaction e . g ., NaNO 2 , HCl, AcOH
  • provides compounds of formula 12-10 which can be decarboxylated ( e .
  • R 2 is via selective cross-coupling with R 2 -M, where M is a boronic acid, boronic ester, or an appropriately substituted metal such as Sn(Bu) 4 or Zn, under standard Suzuki conditions ( e .
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)) or standard Stille conditions ( e.g. , in the presence of a palladium(0) catalyst, such as tetrakis(triphenylphosphine)palladium(0)) or standard Negishi conditions ( e .
  • Ketones 12-13 can be treated with one of the conditions of Scheme 2 to provide an amine that can be converted to compounds of the invention 12-14 according to Scheme 1 .
  • NR 2 can be installed via nucleophilic aromatic substitution of the halogen of 12-12 with amines.
  • Intermediates 12-15 can be treated with one of the conditions of Scheme 2 to provide an amine that can be converted to compounds of the invention 12-16 according to Scheme 1 .
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or bis(triphenylphosphine)palladium(II) chloride and a base ( e.g. , a bicarbonate or carbonate base, or CsF)) or standard Stille conditions ( e.g. , in the presence of a palladium(0) catalyst, such as tetrakis(triphenylphosphine)palladium(0)) or standard Negishi conditions ( e .
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium (II)
  • a palladium catalyst such as tetrakis(triphenylphosphine)palladium(0) or [1,1'-bis(diphenylphosphino)ferrocene] dichloropalladium (II)
  • a suitable oxidant e.g., m -CPBA
  • a suitable reagent e.g. , POX 3 , RCOX, RSO 2 X
  • Protection of the indazole nitrogens can be accomplished to give 1-6 along with its regioisomer. Displacement of the triflate with an appropriate amine can give 1-7 .
  • the ketone in 1-7 can be converted to an amine by various methods as shown in Scheme 3 to furnish amine 1-8 .
  • Amine 1-8 can be coupled with an optionally protected carboxylic acid such as 1-2 ( Scheme 1 ) by various methods as shown in Scheme 1 .
  • any chosen protecting groups can be removed under conditions suitable for their removal, that are also compatible with the functionality present in the resulting compounds of the Formula I and compounds of the invention (compounds of Formula I wherein Y 1 is N, Y 2 is N, Z 1 is C, Z 2 is N). It will be recognized by one skilled in the art that the order of steps in Scheme 14 can be changed in consideration of compatibility of functional groups present in the intermediates.
  • the reactions for preparing compounds described herein can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis.
  • suitable solvents can be substantially non-reactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, (e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature).
  • a given reaction can be carried out in one solvent or a mixture of more than one solvent.
  • suitable solvents for a particular reaction step can be selected by the skilled artisan.
  • ambient temperature and “room temperature” or “rt” as used herein, are understood in the art, and refer generally to a temperature, e.g. a reaction temperature, that is about the temperature of the room in which the reaction is carried out, for example, a temperature from about 20°C to about 30 °C.
  • Preparation of compounds described herein can involve the protection and deprotection of various chemical groups.
  • the need for protection and deprotection, and the selection of appropriate protecting groups, can be readily determined by one skilled in the art.
  • the chemistry of protecting groups can be found, for example, in T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 3rd Ed., Wiley & Sons, Inc., New York (1999 ).
  • Reactions can be monitored according to any suitable method known in the art.
  • product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., 1 H or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV-visible), mass spectrometry, or by chromatographic methods such as high performance liquid chromatography (HPLC), liquid chromatography-mass spectroscopy (LCMS), or thin layer chromatography (TLC).
  • HPLC high performance liquid chromatography
  • LCMS liquid chromatography-mass spectroscopy
  • TLC thin layer chromatography
  • Compounds can be purified by those skilled in the art by a variety of methods, including high performance liquid chromatography (HPLC) and normal phase silica chromatography.
  • the compounds, salts or stereoisomers thereof described herein inhibit activity of PI3K ⁇ kinase. Accordingly, the compounds, salts or stereoisomers described herein can be used in methods of inhibiting PI3K ⁇ kinase by contacting the kinase with any one or more of the compounds, salts, or compositions described herein. In some embodiments, the compounds or salts can be used in methods of inhibiting activity of PI3K ⁇ in an individual in need of said inhibition by administering a inhibiting amount of a compound or salt of described herein. In some embodiments, modulating is inhibiting. In some embodiments, the contacting is in vivo. In some embodiments, the contacting is ex vivo.
  • the PI3K ⁇ includes a mutation.
  • a mutation can be a replacement of one amino acid for another, or a deletion of one or more amino acids.
  • the mutation can be present in the kinase domain of the PI3K ⁇ .
  • the compound or salt further inhibits PI3K ⁇ .
  • the compounds or salts described herein can be selective.
  • selective is meant that the compound binds to or inhibits PI3K ⁇ with greater affinity or potency, respectively, compared to at least one other kinase.
  • the compounds of the invention are selective inhibitors of PI3K ⁇ over PI3K ⁇ , PI3K ⁇ , and PI3K ⁇ .
  • the compounds of the invention are selective inhibitors of PI3K ⁇ over PI3K ⁇ and PI3K ⁇ .
  • selectivity can be at least about 2-fold, 3-fold, 5-fold, 10-fold, at or 20-fold over PI3K ⁇ as measured by the assays described herein.
  • selectivity can be tested at the K m ATP concentration of each enzyme.
  • the selectivity of compounds of the invention can be determined by cellular assays associated with particular PI3K kinase activity.
  • Another aspect of the present invention pertains to compounds of the present invention or a pharmaceutical composition thereof for use in methods of treating a kinase PI3Ky-associated disease or disorder in an individual ( e . g ., patient) by administering to the individual in need of such treatment a therapeutically effective amount or dose of one or more compounds of the present invention or a pharmaceutical composition thereof.
  • a PI3Ky-associated disease or disorder can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of the PI3K ⁇ , including overexpression and/or abnormal activity levels.
  • the disease or disorder is an autoimmune disease or disorder, cancer, cardiovascular disease, or neurodegenerative disease.
  • the disease or disorder is lung cancer (e.g., non-small cell lung cancer), melanoma, pancreatic cancer, breast cancer, prostate cancer, liver cancer, color cancer, endometrial cancer, bladder cancer, skin cancer, cancer of the uterus, renal cancer, gastric cancer, or sarcoma.
  • lung cancer e.g., non-small cell lung cancer
  • melanoma pancreatic cancer
  • breast cancer prostate cancer
  • liver cancer color cancer
  • endometrial cancer bladder cancer
  • skin cancer cancer of the uterus
  • renal cancer gastric cancer
  • sarcoma sarcoma
  • the sarcoma is Askin's tumor, sarcoma botryoides, chondrosarcoma, Ewing's sarcoma, malignant hemangioendothelioma, malignant schwannoma, osteosarcoma, alveolar soft part sarcoma, angiosarcoma, cystosarcoma phyllodes, dermatofibrosarcoma protuberans, desmoid tumor, desmoplastic small round cell tumor, epithelioid sarcoma, extraskeletal chondrosarcoma, extraskeletal osteosarcoma, fibrosarcoma, gastrointestinal stromal tumor (GIST), hemangiopericytoma, hemangiosarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, lymphosarcoma, malignant peripheral nerve sheath tumor (MPNST), neurofibrosarcom
  • the disease or disorder is acute myeloid leukemia (e.g., acute monocytic leukemia), small lymphocyctic lymphoma, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), multiple myeloma, T-cell actute lymphoblasic leukemia (T-ALL), cutaneous T-cell lymphoma, large granular lymphocytic leukemia, mature (peripheral) t-cell neoplasm (PTCL), anaplastic large cell lymphoma (ALCL), or lymphoblastic lymphoma.
  • acute myeloid leukemia e.g., acute monocytic leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • T-ALL T-cell actute lymphoblasic leukemia
  • cutaneous T-cell lymphoma large granular lymphocytic leukemia
  • PTCL mature (
  • the mature (peripheral) t-cell neoplasm is T-cell prolymphocytic leukemia, T-cell granular lymphocytic leukemia, aggressive NK-cell leukemia, mycosis fungoides/Sezary syndrome, naplastic large cell lymphoma (T-cell type), enteropathy type T-cell lymphoma, adult T-cell leukemia/lymphoma, or angioimmunoblastic T-cell lymphoma
  • the anaplastic large cell lymphoma is systemic ALCL or primary cutaneous ALCL.
  • the disease or disorder is Burkitt's lymphoma, acute myeloblastic leukemia, chronic myeloid leukemia, non-Hodgkin's lymphoma, Hodgkin's lymphoma, hairy cell leukemia, Mantle cell lymphoma, small lymphocytic lymphoma, follicular lymphoma, lymphoplasmacytic lymphoma, extranodal marginal zone lymphoma, Waldenstrom's macroglobulinemia, prolymphocytic leukemia, acute lymphoblastic leukemia, myelofibrosis, mucosa-associated lymphatic tissue (MALT) lymphoma, mediastinal (thymic) large B-cell lymphoma, lymphomatoid granulomatosis, splenic marginal zone lymphoma, primary effusion lymphoma, intravascular large B-cell lymphoma, plasma cell leukemia, extramedullary plasmacytoma, smoul
  • the non-Hodgkin's lymphoma is relapsed NHL, refractory NHL, recurrent follicular NHL, indolent NHL (iNHL), or aggressive NHL (aNHL).
  • the diffuse large B cell lymphoma is activated B-cell like (ABC) diffuse large B cell lymphoma, or germinal center B cell (GCB) diffuse large B cell lymphoma.
  • ABSC activated B-cell like
  • GCB germinal center B cell
  • the Burkitt's lymphoma is endemic Burkitt's lymphoma, sporadic Burkitt's lymphoma, or Burkitt's-like lymphoma
  • the disease or disorder is rheumatoid arthritis, multiple sclerosis, systemic lupus erythematous, asthma, allergy, pancreatitis, psoriasis, anaphylaxis, glomerulonephritis, inflammatory bowel disease (e.g., Crohn's disease and ulcerative colitis), thrombosis, meningitis, encephalitis, diabetic retinopathy, benign prostatic hypertrophy, myasthenia gravis, Sjögren's syndrome, osteoarthritis, restenosis, or atherosclerosis.
  • inflammatory bowel disease e.g., Crohn's disease and ulcerative colitis
  • thrombosis e.g., Crohn's disease and ulcerative colitis
  • meningitis e.g., encephalitis
  • diabetic retinopathy e.g., benign prostatic hypertrophy
  • myasthenia gravis e.g., Sjögren's syndrome,
  • disease or disorder is heart hypertropy, cardiac myocyte dysfunction, chronic obstructive pulmonary disease (COPD), elevated blood pressure, ischemia, ischemia-reperfusion, vasoconstriction, anemia (e.g., hemolytic anemia, aplastic anemia, or pure red cell anemia), bacterial infection, viral infection, graft rejection, kidney disease, anaphylactic shock fibrosis, skeletal muscle atrophy, skeletal muscle hypertrophy, angiogenesis, sepsis, graft rejection, glomerulosclerosis, progressive renal fibrosis, idiopathic thrombocytopenic purpura (ITP), autoimmune hemolytic anemia, vasculitis, systemic lupus erythematosus, lupus nephritis, pemphigus, or membranous nephropathy.
  • COPD chronic obstructive pulmonary disease
  • anemia e.g., hemolytic anemia, aplastic anemia, or pure red cell an
  • the disease or disorder is Alzheimer's disease, central nervous system trauma, or stroke.
  • the idiopathic thrombocytopenic purpura is relapsed ITP or refractory ITP.
  • the vasculitis is Behçet's disease, Cogan's syndrome, giant cell arteritis, polymyalgia rheumatica (PMR), Takayasu's arteritis, Buerger's disease (thromboangiitis obliterans), central nervous system vasculitis, Kawasaki disease, polyarteritis nodosa, Churg-Strauss syndrome, mixed cryoglobulinemia vasculitis (essential or hepatitis C virus (HCV)-induced), Henoch-Schonlein purpura (HSP), hypersensitivity vasculitis, microscopic polyangiitis, Wegener's granulomatosis, or anti-neutrophil cytoplasm antibody associated (ANCA) systemic vasculitis (AASV).
  • ANCA anti-neutrophil cytoplasm antibody associated
  • the present invention further provides a compound described herein, or a pharmaceutically acceptable salt thereof, for use in any of the methods described herein.
  • the present invention further provides use of a compound described herein, or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for use in any of the methods described herein.
  • contacting refers to the bringing together of indicated moieties in an in vitro system or an in vivo system.
  • "contacting" a PI3K with a compound of the invention includes the administration of a compound of the present invention to an individual or patient, such as a human, having a PI3K, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the PI3K.
  • the term "individual” or “patient,” used interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.
  • the phrase "therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response that is being sought in a tissue, system, animal, individual or human by a researcher, veterinarian, medical doctor or other clinician.
  • treating can refer to one or more of (1) inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., arresting further development of the pathology and/or symptomatology); and (2) ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e., reversing the pathology and/or symptomatology) such as decreasing the severity of disease.
  • One or more additional pharmaceutical agents such as, for example, chemotherapeutics, antiinflammatory agents, steroids, immunosuppressants, immune-oncology agents, metabolic enzyme inhibitors, chemokine receptor inhibitors, and phosphatase inhibitors, as well as Bcr-Abl, Flt-3, EGFR, HER2, JAK, c-MET, VEGFR, PDGFR, cKit, IGF-1R, RAF and FAK kinase inhibitors such as, for example, those described in WO 2006/056399 .
  • Other agents such as therapeutic antibodies can be used in combination with the compounds of the present invention for treatment of PI3K-associated diseases, disorders or conditions.
  • the one or more additional pharmaceutical agents can be administered to a patient simultaneously or sequentially.
  • the compounds of the present disclosure can be combined with one or more inhibitors of the following kinases for the treatment of cancer: Akt1, Akt2, Akt3, TGF- ⁇ R, PKA, PKG, PKC, CaM-kinase, phosphorylase kinase, MEKK, ERK, MAPK, mTOR, EGFR, HER2, HER3, HER4, INS-R, IGF-1R, IR-R, PDGFaR, PDGF ⁇ R, CSFIR, KIT, FLK-II, KDR/FLK-1, FLK-4, flt-1, FGFR1, FGFR2, FGFR3, FGFR4, c-Met, Ron, Sea, TRKA, TRKB, TRKC, FLT3, VEGFR/Flt2, Flt4, EphA1, EphA2, EphA3, EphB2, EphB4, Tie2, Src, Fyn, Lck, Fgr, Btk, Fak, SYK
  • the compounds of the present disclosure can be combined with one or more of the following inhibitors for the treatment of cancer or infections.
  • inhibitors that can be combined with the compounds of the present disclosure for treatment of cancer and infections include an FGFR inhibitor (FGFR1, FGFR2, FGFR3 or FGFR4, e.g., INCB54828, INCB62079 and INCB63904), a JAK inhibitor (JAK1 and/or JAK2, e.g., ruxolitinib, baricitinib or INCB39110), an IDO inhibitor (e.g., epacadostat and NLG919), an LSD1 inhibitor (e.g., INCB59872 and INCB60003), a TDO inhibitor, a PI3K-delta inhibitor (e.g., INCB50797 and INCB50465), a Pim inhibitor, a CSF1R inhibitor, a TAM receptor tyrosine kinases (Tyro-3,
  • the compound or salt described herein is administered with a PI3K ⁇ inhibitor. In some embodiments, the compound or salt described herein is administered with a JAK inhibitor. In some embodiments, the compound or salt described herein is administered with a JAK1 or JAK2 inhibitor (e.g., baricitinib or ruxolitinib). In some embodiments, the compound or salt described herein is administered with a JAK1 inhibitor. In some embodiments, the compound or salt described herein is administered with a JAK1 inhibitor, which is selective over JAK2.
  • Example antibodies for use in combination therapy include but are not limited to Trastuzumab (e.g. anti-HER2), Ranibizumab (e.g. anti-VEGF-A), Bevacizumab (trade name Avastin, e.g. anti-VEGF, Panitumumab (e.g. anti-EGFR), Cetuximab (e.g. anti-EGFR), Rituxan (anti-CD20) and antibodies directed to c-MET.
  • Trastuzumab e.g. anti-HER2
  • Ranibizumab e.g. anti-VEGF-A
  • Bevacizumab trade name Avastin, e.g. anti-VEGF, Panitumumab (e.g. anti-EGFR), Cetuximab (e.g. anti-EGFR), Rituxan (anti-CD20) and antibodies directed to c-MET.
  • a cytostatic agent cisplatin, doxorubicin, taxotere, taxol, etoposide, irinotecan, camptostar, topotecan, paclitaxel, docetaxel, epothilones, tamoxifen, 5-fluorouracil, methoxtrexate, temozolomide, cyclophosphamide, SCH 66336, R115777, L778,123, BMS 214662, Iressa, Tarceva, antibodies to EGFR, Gleevec TM , intron, ara-C, adriamycin, cytoxan, gemcitabine, Uracil mustard, Chlormethine, Ifosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylenemelamine, Triethylenethiophosphoramine, Busul
  • chemotherapeutics include proteosome inhibitors (e . g ., bortezomib), thalidomide, revlimid, and DNA-damaging agents such as melphalan, doxorubicin, cyclophosphamide, vincristine, etoposide, carmustine, and the like.
  • Example steroids include corticosteroids such as dexamethasone or prednisone.
  • Example Bcr-Abl inhibitors include the compounds, and pharmaceutically acceptable salts thereof, of the genera and species disclosed in U.S. Pat. No. 5,521,184 , WO 04/005281 , and U.S. Ser. No. 60/578,491 .
  • Example suitable Flt-3 inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 03/037347 , WO 03/099771 , and WO 04/046120 .
  • Example suitable RAF inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 00/09495 and WO 05/028444 .
  • Example suitable FAK inhibitors include compounds, and their pharmaceutically acceptable salts, as disclosed in WO 04/080980 , WO 04/056786 , WO 03/024967 , WO 01/064655 , WO 00/053595 , and WO 01/014402 .
  • the compounds of the invention can be used in combination with one or more other kinase inhibitors including imatinib, particularly for treating patients resistant to imatinib or other kinase inhibitors.
  • the compounds of the invention can be used in combination with a chemotherapeutic in the treatment of cancer, and may improve the treatment response as compared to the response to the chemotherapeutic agent alone, without exacerbation of its toxic effects.
  • the compounds of the invention can be used in combination with a chemotherapeutic provided herein.
  • additional pharmaceutical agents used in the treatment of multiple myeloma can include, without limitation, melphalan, melphalan plus prednisone [MP], doxorubicin, dexamethasone, and Velcade (bortezomib).
  • Additional agents used in the treatment of multiple myeloma include Bcr-Abl, Flt-3, RAF and FAK kinase inhibitors. Additive or synergistic effects are desirable outcomes of combining a PI3K inhibitor of the present invention with an additional agent.
  • PI3K ⁇ inhibitors provided herein can be used in combination with one or more immune checkpoint inhibitors for the treatment of cancer as described herein.
  • the combination with one or more immune checkpoint inhibitors as described herein can be used for the treatment of melanoma.
  • Exemplary immune checkpoint inhibitors include inhibitors against immune checkpoint molecules such as CD27, CD28, CD40, CD122, OX40, GITR, CD137, ICOS, A2AR, B7-H3, B7-H4, BTLA, CTLA-4, LAG3, TIM3, VISTA, PD-1, PD-L1 and PD-L2.
  • the compounds provided herein can be used in combination with one or more agents selected from KIR inhibitors, TIGIT inhibitors, LAIR1 inhibitors, CD160 inhibitors, 2B4 inhibitors and TGFR beta inhibitors.
  • the inhibitor of an immune checkpoint molecule is anti-PD 1 antibody, anti-PD-Ll antibody, or anti-CTLA-4 antibody.
  • the inhibitor of an immune checkpoint molecule is a small molecule PD-1 inhibitor or a small molecule PD-L1 inhibitor.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-1, e.g., an anti-PD-1 monoclonal antibody.
  • the anti-PD-1 monoclonal antibody is nivolumab, pembrolizumab (also known as MK-3475), pidilizumab, SHR-1210, or AMP-224.
  • the anti-PD-1 monoclonal antibody is nivolumab or pembrolizumab.
  • the anti-PDl antibody is pembrolizumab.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of PD-L1, e.g., an anti-PD-Ll monoclonal antibody.
  • the anti-PD-Ll monoclonal antibody is BMS-935559, MEDI4736, MPDL3280A (also known as RG7446), or MSB0010718C.
  • the anti-PD-Ll monoclonal antibody is MPDL3280A or MEDI4736.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of CTLA-4, e.g., an anti-CTLA-4 antibody.
  • the anti-CTLA-4 antibody is ipilimumab.
  • the inhibitor of an immune checkpoint molecule is an inhibitor of LAG3, e.g., an anti-LAG3 antibody.
  • the anti-LAG3 antibody is BMS-986016.
  • the compounds of the invention can be used in combination with one or more metabolic enzyme inhibitors.
  • the metabolic enzyme inhibitor is an inhibitor of IDOl, TDO, or arginase. Examples of IDOl inhibitors include epacadostat and NGL919.
  • the compounds of the invention can be used in combination with an inhibitor of JAK or PI3K ⁇ .
  • the JAK inhibitor is selective for JAK1 and JAK1 over JAK3 and TYK2. In some embodiments, the JAK inhibitor is selective for JAK1 over JAK2, JAK3, and TYK2. In some embodiments, the JAK inhibitor inhibit JAK1 preferentially over JAK2 (e.g., have a JAK1/JAK2 IC 50 ratio >1). In some embodiments, the JAK inhibitor is about 10-fold more selective for JAK1 over JAK2.
  • the JAK inhibitor is 3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile.
  • the JAK inhibitor is (3 R )-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile (ruxolitinib; also known as INCB018424).
  • Ruxolitinib has an IC 50 of less than 10 nM at 1 mM ATP (assay D) at JAK1 and JAK2.
  • 3-Cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile and ruxolitinib can be made by the procedure described in US 7,598,257 (Example 67), filed December 12, 2006 .
  • the inhibitor of JAK1 and/or JAK2 is (3R)-3-cyclopentyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl]propanenitrile phosphoric acid salt.
  • the inhibitor of JAK1 and/or JAK2 is 2-(3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-1-(ethylsulfonyl)azetidin-3-yl)acetonitrile (baricitinib; also known as INCB028050).
  • the JAK inhibitor is a compound of Table A, or a pharmaceutically acceptable salt thereof.
  • the compounds in Table 1 are selective JAK1 inhibitors (selective over JAK2, JAK3, and TYK2).
  • the IC 50 s obtained by the method of Assay D at 1 mM ATP. Table A # Prep.
  • the PI3K ⁇ inhibitor can be selective.
  • selective is meant that the compound binds to or inhibits a kinase with greater affinity or potency, respectively, compared to at least one other kinase.
  • the PI3K ⁇ inhibitor is a selective inhibitors of PI3K ⁇ (e.g., over PI3K ⁇ , PI3K ⁇ and PI3K ⁇ ).
  • selectivity can be at least about 2-fold, 5-fold, 10-fold, at least about 20-fold, at least about 50-fold, at least about 100-fold, at least about 200-fold, at least about 500-fold or at least about 1000-fold. Selectivity can be measured by methods routine in the art. In some embodiments, selectivity can be tested at the K m ATP concentration of each enzyme. In some embodiments, the selectivity of compounds described herein can be determined by cellular assays associated with particular PI3K kinase activity.
  • the inhibitor of PI3K ⁇ is a compound shown in Table B.
  • the compounds of Table B have been tested in Assay C and shown to be inhibitors of PI3K ⁇ with the IC 50 s in Table B.
  • Table B # Prep. Name Structure PI3K ⁇ IC 50 (nM) 27 US 2011/0015212 (Example 10) 7-(1-(9H-purin-6-ylamino)ethyl)-6-(3-fluorophenyl)-3 -methyl-5H-thiazolo[3,2-a]pyrimidin-5-one + 28 US 2011/0015212 (Example 15) (S)-7-(1-(9H-purin-6-ylamino)ethyl)-6-(3-fluorophenyl)-3 -methyl-5H-thiazolo[3,2-a]pyrimidin-5-one + 29 US 2013/ 0059835 (Example 269) 4-[1-(4-amino-3-methyl-1H-pyra
  • the inhibitor of PI3K ⁇ is selected from:
  • the inhibitor of PI3K ⁇ is selected from:
  • PI3K ⁇ inhibitors provided herein can be administered in combination with an inhibitor of JAK1 and/or JAK2 or an inhibitor of PI3K ⁇ .
  • the agents can be combined with the present compound in a single or continuous dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.
  • a corticosteroid such as dexamethasone is administered to a patient in combination with the compounds of the invention where the dexamethasone is administered intermittently as opposed to continuously.
  • combinations of the compounds of the invention with other therapeutic agents can be administered to a patient prior to, during, and/or after a bone marrow transplant or stem cell transplant.
  • the compounds of the invention can be administered in the form of pharmaceutical compositions.
  • These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal), oral or parenteral.
  • topical including transdermal, epidermal, ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery
  • pulmonary e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal or intranasal
  • oral or parenteral e.g., by inhal
  • Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal intramuscular or injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration.
  • Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump.
  • Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • compositions which contain, as the active ingredient, the compound of the invention or a pharmaceutically acceptable salt thereof, in combination with one or more pharmaceutically acceptable carriers (excipients).
  • the composition is suitable for topical administration.
  • the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container.
  • the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
  • the compounds of the invention may be milled using known milling procedures such as wet milling to obtain a particle size appropriate for tablet formation and for other formulation types.
  • Finely divided (nanoparticulate) preparations of the compounds of the invention can be prepared by processes known in the art, e.g., see International App. No. WO 2002/000196 .
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents.
  • the compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
  • compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 1000 mg (1 g), more usually about 100 to about 500 mg, of the active ingredient.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • compositions of the invention contain from about 5 to about 50 mg of the active ingredient.
  • the compositions of the invention contain from about 5 to about 50 mg of the active ingredient.
  • One having ordinary skill in the art will appreciate that this embodies compositions containing about 5 to about 10, about 10 to about 15, about 15 to about 20, about 20 to about 25, about 25 to about 30, about 30 to about 35, about 35 to about 40, about 40 to about 45, or about 45 to about 50 mg of the active ingredient.
  • compositions of the invention contain from about 50 to about 500 mg of the active ingredient.
  • the compositions of the invention contain from about 500 to about 1000 mg of the active ingredient.
  • One having ordinary skill in the art will appreciate that this embodies compositions containing about 500 to about 550, about 550 to about 600, about 600 to about 650, about 650 to about 700, about 700 to about 750, about 750 to about 800, about 800 to about 850, about 850 to about 900, about 900 to about 950, or about 950 to about 1000 mg of the active ingredient.
  • the active compound can be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, about 0.1 to about 1000 mg of the active ingredient of the present invention.
  • the tablets or pills of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • liquid forms in which the compounds and compositions of the present invention can be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face mask, tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.
  • Topical formulations can contain one or more conventional carriers.
  • ointments can contain water and one or more hydrophobic carriers selected from, for example, liquid paraffin, polyoxyethylene alkyl ether, propylene glycol, white Vaseline, and the like.
  • Carrier compositions of creams can be based on water in combination with glycerol and one or more other components, e.g. glycerinemonostearate, PEG-glycerinemonostearate and cetylstearyl alcohol.
  • Gels can be formulated using isopropyl alcohol and water, suitably in combination with other components such as, for example, glycerol, hydroxyethyl cellulose, and the like.
  • topical formulations contain at least about 0.1, at least about 0.25, at least about 0.5, at least about 1, at least about 2, or at least about 5 wt % of the compound of the invention.
  • the topical formulations can be suitably packaged in tubes of, for example, 100 g which are optionally associated with instructions for the treatment of the select indication, e.g., psoriasis or other skin condition.
  • compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like.
  • compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.
  • the therapeutic dosage of a compound of the present invention can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician.
  • the proportion or concentration of a compound of the invention in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration.
  • the compounds of the invention can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 ⁇ g/kg to about 1 g/kg of body weight per day.
  • the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day.
  • the dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • compositions of the invention can further include one or more additional pharmaceutical agents such as a chemotherapeutic, steroid, anti-inflammatory compound, or immunosuppressant, examples of which are listed herein.
  • additional pharmaceutical agents such as a chemotherapeutic, steroid, anti-inflammatory compound, or immunosuppressant, examples of which are listed herein.
  • Another aspect of the present invention relates to labeled compounds of the invention (radio-labeled, fluorescent-labeled, etc.) that would be useful not only in imaging techniques but also in assays, both in vitro and in vivo , for localizing and quantitating PI3K in tissue samples, including human, and for identifying PI3K ligands by inhibition binding of a labeled compound.
  • the present invention includes PI3K assays that contain such labeled compounds.
  • the present invention further includes isotopically-labeled compounds of the invention.
  • An “isotopically” or “radio-labeled” compound is a compound of the invention where one or more atoms are replaced or substituted by an atom having an atomic mass or mass number different from the atomic mass or mass number typically found in nature ( i.e. , naturally occurring).
  • Suitable radionuclides that may be incorporated in compounds of the present invention include but are not limited to 2 H (also written as D for deuterium), 3 H (also written as T for tritium), 11 C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 18 F, 35 S, 36 C1, 82 Br, 75 Br, 76 Br, 77 Br, 123 I, 124 I, 125 I and 131 I.
  • the radionuclide that is incorporated in the instant radio-labeled compounds will depend on the specific application of that radio-labeled compound.
  • a “radio-labeled” or “labeled compound” is a compound that has incorporated at least one radionuclide.
  • the radionuclide is selected from the group consisting of 3 H, 14 C, 125 I, 35 S and 82 Br.
  • the present invention can further include synthetic methods for incorporating radio-isotopes into compounds of the invention. Synthetic methods for incorporating radio-isotopes into organic compounds are well known in the art, and an ordinary skill in the art will readily recognize the methods applicable for the compounds of invention.
  • a labeled compound of the invention can be used in a screening assay to identify/evaluate compounds.
  • a newly synthesized or identified compound i.e. , test compound
  • a test compound which is labeled can be evaluated for its ability to bind a PI3K by monitoring its concentration variation when contacting with the PI3K, through tracking of the labeling.
  • a test compound (labeled) can be evaluated for its ability to reduce binding of another compound which is known to bind to a PI3K (i.e. , standard compound). Accordingly, the ability of a test compound to compete with the standard compound for binding to the PI3K directly correlates to its binding affinity.
  • the standard compound is labeled and test compounds are unlabeled. Accordingly, the concentration of the labeled standard compound is monitored in order to evaluate the competition between the standard compound and the test compound, and the relative binding affinity of the test compound is thus ascertained.
  • kits useful for example, in the treatment or prevention of PI3K-associated diseases or disorders, such as cancer, which include one or more containers containing a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention.
  • kits can further include, if desired, one or more of various conventional pharmaceutical kit components, such as, for example, containers with one or more pharmaceutically acceptable carriers, additional containers, etc., as will be readily apparent to those skilled in the art.
  • Instructions, either as inserts or as labels, indicating quantities of the components to be administered, guidelines for administration, and/or guidelines for mixing the components, can also be included in the kit.
  • pH 10 purifications: Waters XBridge C 18 5 ⁇ m, 19 x 100 mm column, eluting with mobile phase A: 0.15% NH 4 OH in water and mobile phase B: acetonitrile; the flow rate was 30 mL/minute, the separating gradient was optimized for each compound using the Compound Specific Method Optimization protocol as described in the literature (see e.g. " Preparative LCMS Purification: Improved Compound Specific Method Optimization", K. Blom, B. Glass, R. Sparks, A. Combs, J. Comb. Chem., 6, 874-883 (2004 )). Typically, the flow rate used with 30 x 100 mm column was 60 mL/minute.
  • Step 1 tert-butyl 3-(1-(4-chloro-7-ethoxy-1-methyl-1H-indazol-6-yl)ethylcarbamoyl)pyrazolo[1,5-a]pyrimidin-2-ylcarbamate & tert-butyl 3-(1-(4-chloro-7-ethoxy-2-methyl-2H-indazol-6-yl)ethylcarbamoyl)pyrazolo[1,5-a]pyrimidin-2-ylcarbamate
  • Examples 5-13 were synthesized via an alkylation with the appropriate alkyl halide and subsequent deprotection as described for Example 4. In each case, the alkylation step afforded a varying mixture of N1 and N2 indazole regioisomers. These isomers were separable by preparative HPLC after deprotection, unless otherwise noted.
  • the N1 regioisomers prepared and the corresponding data are listed in Table 1.
  • the N2 regioisomers prepared and the corresponding data are listed in Table 2. Table 1. Ex. No.
  • Trifluoromethanesulfonic anhydride (1M/CH 2 Cl 2 , 13 mL, 13 mmol) was added to a solution of 1-(5-chloro-2-hydroxy-4-methyl-3-nitrophenyl)ethanone (2.0 g, 8.7 mmol, from Oakwood) and triethylamine (2.4 mL, 17 mmol) in THF (20 mL) at -78 °C. The solution was allowed to warm to room temperature and stirred for 0.5 h. The reaction mixture was diluted with EtOAc (30 mL) and quenched with sat. NaHCO 3 (20 mL). The layers were separated and the organic layer was washed with sat.
  • Step 1 tert-butyl 3-(1-(4-chloro-1-methyl-7-phenyl-1H-indazol-6-yl)ethylcarbamoyl)pyrazolo[1,5-a]pyrimidin-2-ylcarbamate & tert-butyl 3-(1-(4-chloro-2-methyl-7-phenyl-2H-indazol-6-yl)ethylcarbamoyl)pyrazolo[1,5-a]pyrimidin-2-ylcarbamate
  • Step 1 tert-butyl 3-(1-(4-chloro-2-(2-(1,3-dioxoisoindolin-2-yl)ethyl)-7-phenyl-2H-indazol-6- yl)ethylcarbamoyl)pyrazolo[1,5-a]pyrimidin-2-ylcarbamate & tert-butyl 3-(1-(4-chloro-1-(2-(1,3-dioxoisoindolin-2-yl)ethyl)-7-phenyl-1H-indazol-6-yl)ethylcarbamoyl)pyrazolo[1,5-a]pyrimidin-2-ylcarbamate
  • Triethylamine (16.6 mL, 119 mmol) and trimethylsilyl cyanide (25.9 mL, 194 mmol) were added to a solution of methyl 2,5-dichloronicotinate 1-oxide (17.4 g, 78.4 mmol, from Step 2) in acetonitrile (150 mL).
  • the reaction mixture was heated to 70 °C for 20 minutes. Upon cooling to room temperature, the reaction mixture was diluted with EtOAc, and the solution was quenched by slow addition to a cold solution of aqueous K 2 CO 3 (500 mL). The resulting aqueous mixture was extracted with DCM.
  • the scalemic mixture was separated by HPLC (Phenomenex Lux Cellulose C-1, 5 ⁇ m, 21.2 x 250 mm, 9 mg/900 ⁇ L loading, eluting with 20% EtOH in hexanes at 18 mL/min over 13 min).
  • HPLC Henomenex Lux Cellulose C-1, 5 ⁇ m, 21.2 x 250 mm, 9 mg/900 ⁇ L loading, eluting with 20% EtOH in hexanes at 18 mL/min over 13 min).
  • Titanium tetraisopropoxide (0.052 mL, 0.18 mmol) was added to a mixture of 1-[5-(3-fluorophenyl)-3,8-dimethylimidazo[1,5- a ]pyridin-6-yl]ethanone (0.025 g, 0.088 mmol, prepared as in Step 1) in 2.0 M ammonia in ethanol (0.22 mL, 0.44 mmol). The reaction was heated to 60 °C for 2 hours. The reaction mixture was then cooled to 0 °C, and NaBH 4 (0.0050 g, 0.13 mmol) was added. After 30 minutes, the reaction mixture was quenched with water and insoluble material was removed by filtration.
  • a microwavable vial was charged with 1-(8-chloro-5-phenylimidazo[1,5-a]pyridin-6-yl)ethanone (50. mg, 0.18 mmol, from Example 27, Step 7), Zn(CN) 2 (24 mg, 0.20 mmol), tris(dibenzylideneacetone)dipalladium(0) chloroform adduct (11 mg, 0.011 mmol), and 2-(dicyclohexylphosphino)-2',6'-dimethoxy-1,1'-biphenyl (7.6 mg, 0.018 mmol). DMF (3.35 mL) and water (17 ⁇ L, 0.92 mmol) were added.
  • 6-Acetyl-5-phenylimidazo[1,5-a]pyridine-8-carbonitrile (36 mg, 0.14 mmol, from Step 1) was dissolved in MeOH (1.5 mL) and ammonium acetate (106 mg, 1.38 mmol) was added. The resulting mixture was heated at 65 °C for 1 hour. Sodium cyanoborohydride (26 mg, 0.41 mmol) was added and the solution was heated at 65 °C ovrnight. Upon cooling to room temperature, the reaction was quenched by the addition of water was added.
  • Examples 31A-31B 2-Amino-N-((1S)-1-(8-chloro-5-(2-methyl-1,1-dioxidothiomorpholino)imidazo[1,5-a]pyridin-6-yl)ethyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide trifluoroacetate salt (Isomers 1-2, scalemic mixture prepared)
  • N , N -Diethylpropan-2-amine (2.45 ml, 15.8 mmol) was added to 2-(( tert- butoxycarbonyl)amino)pyrazolo[1,5- a ]pyrimidine-3-carboxylic acid (1.32 g, 4.7 mmol) and N,N,N', N '-tetratnethyl-O-(7-azabenzotriazol-1-yl)uronium hexafluorophosphate (1.65 g, 4.3 mmol) in N,N- dimethylformamide (8 mL).
  • Isomer 2 was synthesized according to steps 3-6, using single isomer 2 from step 2 as starting material for step 3.
  • Trifluoromethanesulfonic anhydride (2 mL, 9 mmol) was added dropwise to ethyl 2-amino-5-oxo-4,5-dihydropyrazolo[1,5- a ]pyrimidine-3-carboxylate (0.7 g, 3 mmol) in dichloromethane (20 mL) and triethylamine (3 mL, 20 mmol) at -78 °C. The resulting mixture was stirred at -78 °C for 1 h. The reaction was quenched by adding sodium bicarbonate (sat.) at -78°C. The mixture was futher diluted with dichloromethane (10 mL) and stirred from -78°C to room temperature.
  • This compound was synthesized according to the procedure of Example 41, using 1-(3,4-dichloro-7-ethoxy-1 H -indazol-6-yl)ethanamine to replace 1-(4-chloro-7-ethoxy-1 H -indazol-6-yl)ethanamine as the starting material in step 4.
  • 6-chloro-3-iodoimidazo[1,2- b ]pyridazin-2-amine 300 mg, 1.0 mmol
  • methanol (20 mL) and triethylamine 0.52 mL, 3.8 mmol
  • triethylamine 0.52 mL, 3.8 mmol
  • dichloromethane 1:1
  • the solution was saturated with carbon monoxide by bubbling the carbon monoxide gas through the reaction subsurface for 3 min.
  • the vessel was sealed and heated to 55°C for 12 h.
  • Example 45 This compound was synthesized according to the procedure of Example 44, using methyl 2-amino-6-chloroimidazo[1,2- b ]pyridazine-3-carboxylate (Example 45, Step 2) to replace ethyl 2-amino-5- ⁇ [(trifluoromethyl)sulfonyl]oxy ⁇ pyrazolo[1,5- a ]pyrimidine-3-carboxylate as one of the starting materials in step 1.
  • LCMS calculated for C 23 H 28 ClN 8 O 2 (M+H) + : m/z 483.2; found: 483.2.
  • Step 4 2-amino-N-(1-(3-bromo-4-chloro-7-ethoxy-1H-indazol-6-yl)ethyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide trifluoroacetate
  • This compound was synthesized according to the procedure described in Example 1, Step 7, starting from tert -butyl 3-(1-(3-bromo-4-chloro-7-ethoxy-1H-indazol-6-yl)ethylcarbamoyl)pyrazolo[1,5-a]pyrimidin-2-ylcarbamate (10 mg, 0.02 mmol). The residue was purified by preparative HPLC (pH 2) to afford the product as a white solid.
  • Examples 53-54 2-Amino- N -(1-(3-bromo-4-chloro-7-ethoxy-2-(2-hydroxyethyl)-2 H -indazol-6-yl)ethyl)pyrazolo[1,5- a ]pyrimidine-3-carboxamide (Example 53) & 2-Amino- N -(1-(3-bromo-4-chloro-7-ethoxy-1-(2-hydroxyethyl)-1 H -indazol-6-yl)ethyl)pyrazolo[1,5- a ]pyrimidine-3-carboxamide trifluoroacetate (Example 54)
  • Step 7 2-amino-N-(1-(4-chloro-3-cyano-7-phenyl-1H-indazol-6-yl)ethyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide trifluoroacetate
  • Ethyl 6-acetyl-7-amino-4-chloropyrazolo[1,5-a]pyridine-3-carboxylate (413 mg, 1.47 mmol) was dissolved in conc. HCl (6.0 mL) and acetic acid (6.0 mL) and cooled to 0 °C.
  • a solution of sodium nitrite (300 mg, 4.4 mmol) in water (1.0 mL) was added dropwise, and the solution changed from yellow to dark green. After 0.5 h, the reaction mixture was diluted with water and EtOAc, and the layers were separated. The organic layer was washed with brine, dried over MgSO 4 , and concentrated.
  • Example 57-58 ( S )-2-Amino- N -(1-(4-chloro-7-(1,1-dioxidothiomorpholino)pyrazolo[1,5- a ]pyridin-6-yl)ethyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide trifluoroacetate (Example 57) & ( S )-2-Amino- N -(1-(4-chloro-7-(1,1-dioxidothiomorpholino)-3-fluoropyrazolo[1,5- a ]pyridin-6-yl)ethyl)pyrazolo[1,5-a]pyrimidine-3-carboxamide trifluoroacetate (Example 58)
  • Step 2 1-(4-chloro-7-(1,1-dioxidothiomorpholino)pyrazolo[1,5-a]pyridin-6-yl)ethan-1-one & 1-(4-chloro-7-(1,1-dioxidothiomorpholino)-3-fluoropyrazolo[1,5-a]pyridin-6-yl)ethan-1-one
  • Step 3 N-((S)-1-(4-chloro-7-(1,1-dioxidothiomorpholino)pyrazolo[1,5-a]pyridin-6-yl)ethyl)-2-methylpropane-2-sulfinamide & N-((S)-1-(4-chloro-7-(1,1-dioxidothiomorpholino)-3-fluoropyrazolo[1,5-a]pyridin-6-yl)ethyl)-2-methylpropane-2-sulfinamide
  • the resulting mixture of sulfinimines was cooled to 0 °C and sodium borohydride (1.189 g, 31.4 mmol) was added in several portions.
  • the reaction mixture was allowed to warm to room temperature and a small amount of EtOH ( ⁇ 0.5 mL) was added dropwise. After stirring for 0.5 h, LCMS indicated complete conversion (6:1 d.r. for both compounds).
  • the reaction mixture was cooled to 0 °C, quenched by the dropwise addition of MeOH, and vigorously stirred until no more gas evolution was observed.
  • the solution was poured into brine and the resulting suspension was filtered through celite. The filtrate was diluted with EtOAc and water, and the layers were separated.
  • Example 60 2-Amino- N -(1-(4-chloro-3-cyano-7-(1,1-dioxidothiomorpholino)pyrazolo[1,5- a ]pyridin-6-yl)ethyl)pyrazolo[1,5- a ]pyrimidine-3-carboxamide trifluoroacetate
  • This compound was prepared according to the procedure described in Example 56, Step 9, starting from 1-(8-chloro-5-cyclopentylimidazo[1,5-a]pyridin-6-yl)ethanone.
  • the product was purified by preparative HPLC (pH 2) to afford the title compound as a white solid.
  • This compound was prepared according to the procedure described in Example 56, Step 9, starting from 1-(4-chloro-7-phenyl-[1,2,3]triazolo[1,5-a]pyridin-6-yl)ethanone.
  • the product was purified by preparative HPLC (pH 2).
  • Step 3 tert-Butyl ⁇ 3-[(6-acetyl-4-chloro-1H-indazol-7-yl)oxy]propyl]carbamate
  • Step 4 tert-Butyl (3- ⁇ [6-(1-aminoethyl)-4-chloro-1H-indazol-7-yl]oxy ⁇ propyl)carbamate
  • Step 5 tert-Butyl [3-( ⁇ [1-(7- ⁇ 3-[(tert-butoxycarbonyl)amino]propoxy ⁇ -4-chloro-1H-indazol-6-yl)ethyl]amino ⁇ carbonyl)pyrazolo[1,5-a]pyrimidin-2-yl]carbamate
  • Step 6 2-Amino-N- ⁇ 1-[7-(3-aminopropoxy)-4-chloro-1H-indazol-6-yl)ethyl ⁇ pyrazolo[1,5-a]pyrimidine-3-carboxamide bis(trifluoroacetate)
  • the reaction mixture was then cooled to 0 °C, and sodium borohydride (6 mg, 0.1 mmol) was added. The reaction mixture was stirred overnight after coming to room temperature. The reaction mixture was again cooled to 0 °C, and the reaction was quenched with MeOH (0.1 mL). The mixture was warmed to room temperature and diluted with EtOAC (0.25 mL). Brine (10 ⁇ L) was then added, and the resulting slurry was stirred for 5 min. The slurry was filtered through Celite, and the filter cake was washed with EtOAc. The filtrate was concentrated.
  • Diisopropyl azodicarboxylate (0.26 mL, 1.3 mmol) was added dropwise to a mixture of 1-(5-chloro-2-hydroxy-4-methyl-3-nitrophenyl)ethanone (0.15 g, 0.65 mmol, from Biogene Organics, BG-C1031), 2- ⁇ [ tert -butyl(dimethyl)silyl]oxy ⁇ ethanol (0.26 mL, 1.3 mmol), and triphenylphosphine (0.39 g, 1.5 mmol) in THF at 0 °C. The 0 °C bath was removed, and the reaction mixture was stirred for 6 h at room temperature. The reaction mixture was partitioned between water and EtOAc.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Transplantation (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Dermatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Psychiatry (AREA)
  • Hospice & Palliative Care (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
EP16805238.9A 2015-11-06 2016-11-04 Heterocyclic compounds as pi3k-gamma inhibitors Active EP3371190B1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
RS20220521A RS63359B1 (sr) 2015-11-06 2016-11-04 Heterociklična jedinjenja kao inhibitori pi3k-gama
HRP20220599TT HRP20220599T1 (hr) 2015-11-06 2016-11-04 Heterociklički spojevi kao inhibitori pi3k-gama
EP22162035.4A EP4086259A1 (en) 2015-11-06 2016-11-04 Heterocyclic compounds as pi3k-gamma inhibitors
SI201631549T SI3371190T1 (sl) 2015-11-06 2016-11-04 Heterociklične spojine kot inhibitorji PI3K gama

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562252050P 2015-11-06 2015-11-06
PCT/US2016/060468 WO2017079519A1 (en) 2015-11-06 2016-11-04 Heterocyclic compounds as pi3k-gamma inhibitors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP22162035.4A Division EP4086259A1 (en) 2015-11-06 2016-11-04 Heterocyclic compounds as pi3k-gamma inhibitors

Publications (2)

Publication Number Publication Date
EP3371190A1 EP3371190A1 (en) 2018-09-12
EP3371190B1 true EP3371190B1 (en) 2022-03-30

Family

ID=57460598

Family Applications (2)

Application Number Title Priority Date Filing Date
EP22162035.4A Withdrawn EP4086259A1 (en) 2015-11-06 2016-11-04 Heterocyclic compounds as pi3k-gamma inhibitors
EP16805238.9A Active EP3371190B1 (en) 2015-11-06 2016-11-04 Heterocyclic compounds as pi3k-gamma inhibitors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP22162035.4A Withdrawn EP4086259A1 (en) 2015-11-06 2016-11-04 Heterocyclic compounds as pi3k-gamma inhibitors

Country Status (17)

Country Link
US (4) US10065963B2 (es)
EP (2) EP4086259A1 (es)
JP (1) JP6854817B2 (es)
AR (1) AR106595A1 (es)
DK (1) DK3371190T3 (es)
ES (1) ES2915550T3 (es)
HR (1) HRP20220599T1 (es)
HU (1) HUE059324T2 (es)
LT (1) LT3371190T (es)
MA (1) MA43169B1 (es)
MD (1) MD3371190T2 (es)
PL (1) PL3371190T3 (es)
PT (1) PT3371190T (es)
RS (1) RS63359B1 (es)
SI (1) SI3371190T1 (es)
TW (2) TWI824309B (es)
WO (1) WO2017079519A1 (es)

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3215509B1 (en) 2014-11-06 2020-02-26 Lysosomal Therapeutics Inc. Substituted pyrrolo[1,2-a]pyrimidines and their use in the treatment of medical disorders
US20170333435A1 (en) 2014-11-06 2017-11-23 Lysosomal Therapeutics Inc. Substituted imidazo[1,5-a]pyrimidines and their use in the treatment of medical disorders
FI3215511T3 (fi) 2014-11-06 2024-05-16 Bial R&D Invest S A Substituoituja pyratsolo(1,5-a)pyrimidiinejä ja niiden käyttö lääketieteellisten häiriöiden hoitamisessa
WO2016130501A1 (en) 2015-02-09 2016-08-18 Incyte Corporation Aza-heteroaryl compounds as pi3k-gamma inhibitors
TWI824309B (zh) 2015-11-06 2023-12-01 美商英塞特公司 作為PI3K-γ抑制劑之雜環化合物
US20170190689A1 (en) 2016-01-05 2017-07-06 Incyte Corporation Pyridine and pyridimine compounds as pi3k-gamma inhibitors
EP3419971B1 (en) * 2016-02-25 2022-04-20 Asceneuron SA Glycosidase inhibitors
EP3440080A4 (en) 2016-04-06 2020-01-22 Lysosomal Therapeutics Inc. PYRAZOLO [1,5-A] PYRIMIDINYL CARBOXAMIDE COMPOUNDS AND THEIR USE IN THE TREATMENT OF MEDICAL DISORDERS
EP3440081A4 (en) 2016-04-06 2019-09-18 Lysosomal Therapeutics Inc. PYRROLO [1,2-A] PYRIMIDINYL CARBOXAMIDE COMPOUNDS AND THEIR USE IN THE TREATMENT OF MEDICAL DISORDERS
MX2018012208A (es) 2016-04-06 2019-03-28 Lysosomal Therapeutics Inc Compuestos a base de imidazo [1,5-a] pirimidinil carboxamida y su uso en el tratamiento de trastornos médicos.
WO2017192931A1 (en) 2016-05-05 2017-11-09 Lysosomal Therapeutics Inc. SUBSTITUTED IMDAZO[1,2-α]PYRIDINES, SUBSTITUTED IMIDAZO[1,2-α]PYRAZINES, RELATED COMPOUNDS, AND THEIR USE IN THE TREATMENT OF MEDICAL DISORDERS
US11168087B2 (en) 2016-05-05 2021-11-09 Bial—R&D Investments, S.A. Substituted imidazo[1,2-b]pyridazines, substituted imidazo[1,5-b]pyridazines, related compounds, and their use in the treatment of medical disorders
WO2017223414A1 (en) 2016-06-24 2017-12-28 Incyte Corporation HETEROCYCLIC COMPOUNDS AS PI3K-γ INHIBITORS
MA50398B1 (fr) 2017-10-18 2021-10-29 Incyte Corp Dérivés d'imidazole condensés, substitués par des groupes hydroxy tertiaires, utilisés comme inhibiteurs de pi3k-gamma
TW201927777A (zh) * 2017-12-19 2019-07-16 大陸商基石藥業(蘇州)有限公司 Ido抑制劑
SG11202102224UA (en) 2018-09-05 2021-04-29 Incyte Corp Crystalline forms of a phosphoinositide 3-kinase (pi3k) inhibitor
CA3134814A1 (en) 2019-03-26 2020-10-01 Ventyx Biosciences, Inc. Tyk2 pseudokinase ligands
MX2021012417A (es) * 2019-04-11 2021-11-12 Janssen Pharmaceutica Nv Anillos de piridina que contienen derivados como inhibidores de malt1.
EP3980406A4 (en) * 2019-06-06 2023-06-28 Aligos Therapeutics, Inc. Heterocyclic compounds
WO2021092246A1 (en) * 2019-11-08 2021-05-14 Ventyx Biosciences, Inc. Tyk2 pseudokinase ligands
CN116348453A (zh) * 2021-04-25 2023-06-27 烨辉医药科技(上海)有限公司 杂芳族甲酰胺化合物及其用途

Family Cites Families (89)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4269846A (en) 1979-10-29 1981-05-26 Usv Pharmaceutical Corporation Heterocyclic compounds useful as anti-allergy agents
US5521184A (en) 1992-04-03 1996-05-28 Ciba-Geigy Corporation Pyrimidine derivatives and processes for the preparation thereof
ES2342240T3 (es) 1998-08-11 2010-07-02 Novartis Ag Derivados de isoquinolina con actividad que inhibe la angiogenia.
US6133031A (en) 1999-08-19 2000-10-17 Isis Pharmaceuticals Inc. Antisense inhibition of focal adhesion kinase expression
GB9905075D0 (en) 1999-03-06 1999-04-28 Zeneca Ltd Chemical compounds
GB0004890D0 (en) 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
GB0011092D0 (en) 2000-05-08 2000-06-28 Black James Foundation Gastrin and cholecystokinin receptor ligands (III)
KR100786927B1 (ko) 2000-06-28 2007-12-17 스미스클라인비이참피이엘시이 습식 분쇄방법
MXPA04002243A (es) 2001-09-19 2004-06-29 Aventis Pharma Sa Compuestos quimicos.
EP1441725A1 (en) 2001-10-26 2004-08-04 Aventis Pharmaceuticals Inc. Benzimidazoles and analogues and their use as protein kinases inhibitors
FR2831536A1 (fr) 2001-10-26 2003-05-02 Aventis Pharma Sa Nouveaux derives de benzimidazoles, leur procede de preparation, leur application a titre de medicament, compositions pharmaceutiques et nouvelle utilisation notamment comme inhibiteurs de kdr
CN1582150B (zh) 2001-10-30 2011-09-07 诺瓦提斯公司 作为flt3受体酪氨酸激酶活性抑制剂的星形孢菌素衍生物
DE10207843A1 (de) 2002-02-15 2003-09-04 Schering Ag Mikrolia-Inhibitoren zur Unterbrechung von Interleukin 12 und IFN-gamma vermittelten Immunreaktionen
PE20040522A1 (es) 2002-05-29 2004-09-28 Novartis Ag Derivados de diarilurea dependientes de la cinasa de proteina
GB0215676D0 (en) 2002-07-05 2002-08-14 Novartis Ag Organic compounds
AR042052A1 (es) 2002-11-15 2005-06-08 Vertex Pharma Diaminotriazoles utiles como inhibidores de proteinquinasas
UA80767C2 (en) 2002-12-20 2007-10-25 Pfizer Prod Inc Pyrimidine derivatives for the treatment of abnormal cell growth
US7186832B2 (en) 2003-02-20 2007-03-06 Sugen Inc. Use of 8-amino-aryl-substituted imidazopyrazines as kinase inhibitors
US8524899B2 (en) 2003-03-04 2013-09-03 California Institute Of Technology Alternative heterocycles for DNA recognition
GB0305929D0 (en) 2003-03-14 2003-04-23 Novartis Ag Organic compounds
KR20060054410A (ko) 2003-08-01 2006-05-22 제네랩스 테크놀로지스, 인코포레이티드 플라비비리다에에 대한 2고리 이미다졸 유도체
PE20050952A1 (es) 2003-09-24 2005-12-19 Novartis Ag Derivados de isoquinolina como inhibidores de b-raf
WO2005118580A2 (en) 2004-05-12 2005-12-15 The Government Of The United States Of America As Represented By The Secretary, Department Of Health Tricyclic compounds as inhibitors of the hypoxic signaling pathway
AU2005309019A1 (en) 2004-11-24 2006-06-01 Novartis Ag Combinations of JAK inhibitors and at least one of Bcr-Abl, Flt-3, FAK or RAF kinase inhibitors
ES2431050T3 (es) 2005-08-04 2013-11-22 Sirtris Pharmaceuticals, Inc. Benzotiazoles y tiazolopiridinas como moduladores de la sirtuína
JP2009507032A (ja) 2005-09-02 2009-02-19 アボット・ラボラトリーズ 新規なイミダゾ系複素環
PT1945632E (pt) 2005-11-08 2013-12-24 Vertex Pharma Moduladores heterocíclicos de transportadores de cassete de ligação a atp
DK3184526T3 (en) 2005-12-13 2019-01-14 Incyte Holdings Corp PYRROLO [2,3-D] PYRIMIDINE DERIVATIVES AS A JANUS-KINASE INHIBITOR
CN101472912A (zh) 2006-06-22 2009-07-01 比奥维特罗姆上市公司 作为mnk激酶抑制剂的吡啶和吡嗪衍生物
WO2009005551A2 (en) 2007-03-27 2009-01-08 Paratek Pharmaceuticals, Inc. Transcription factor modulating compounds and methods of use thereof
DE102007035333A1 (de) 2007-07-27 2009-01-29 Boehringer Ingelheim Pharma Gmbh & Co. Kg Neue substituierte Arylsulfonylglycine, deren Herstellung und deren Verwendung als Arzneimittel
GB0716292D0 (en) 2007-08-21 2007-09-26 Biofocus Dpi Ltd Imidazopyrazine compounds
US20110052562A1 (en) 2007-12-19 2011-03-03 The Scripps Research Institute Benzimidazoles and analogs as rho kinase inhibitors
US8623347B2 (en) 2008-01-15 2014-01-07 Siga Technologies, Inc. Antiviral drugs for treatment of arenavirus infection
AR071523A1 (es) 2008-04-30 2010-06-23 Merck Serono Sa Compuestos biciclicos fusionados, un proceso para su preparacion, el compuesto para ser utilizado como medicamento en el tratamiento y profilaxis de enfermedades, una composicion farmaceutica y un conjunto que comprende paquetes separados del compuesto y de un ingrediente activo del medicamento
CA2763536C (en) 2008-05-30 2017-05-09 Marvin J. Miller Anti-bacterial agents from benzo[d]heterocyclic scaffolds for prevention and treatment of multidrug resistant bacteria
US8470819B2 (en) 2008-11-03 2013-06-25 Merck Sharp & Dohme Corp. Benzimidazole and aza-benzimidazole carboxamides
TW201028399A (en) 2008-11-27 2010-08-01 Shionogi & Co Pyrimidine derivative and pyridine derivative both having pi3k inhibitory activity
GB0822981D0 (en) 2008-12-17 2009-01-21 Summit Corp Plc Compounds for treatment of duchenne muscular dystrophy
CA2753560A1 (en) 2009-02-27 2010-11-25 Vertex Pharmaceuticals Incorporated Inhibitors of phosphatidylinositol 3-kinase
KR101771401B1 (ko) 2009-05-22 2017-08-25 인사이트 홀딩스 코포레이션 야누스 키나제 억제제로서 피라졸­4­일­피롤로[2,3­d]피리미딘 및 피롤­3­일­피롤로[2,3­d]피리미딘의 N­(헤테로)아릴­피롤리딘 유도체
MY162507A (en) 2009-06-29 2017-06-15 Incyte Holdings Corp Pyrimidinones as pi3k inhibitors
US9249145B2 (en) 2009-09-01 2016-02-02 Incyte Holdings Corporation Heterocyclic derivatives of pyrazol-4-yl-pyrrolo[2,3-d]pyrimidines as janus kinase inhibitors
US8680108B2 (en) 2009-12-18 2014-03-25 Incyte Corporation Substituted fused aryl and heteroaryl derivatives as PI3K inhibitors
US8759359B2 (en) 2009-12-18 2014-06-24 Incyte Corporation Substituted heteroaryl fused derivatives as PI3K inhibitors
WO2011099832A2 (en) 2010-02-12 2011-08-18 Crystalgenomics, Inc. Novel benzimidazole compound, preparation method thereof and pharmaceutical composition comprising the same
SI3050882T1 (en) 2010-03-10 2018-06-29 Incyte Holdings Corporation Piperidin-4-yl azetidine derivatives, as inhibitors of JAK1
WO2011123609A1 (en) 2010-03-31 2011-10-06 Glaxo Group Limited Imidazolyl-imidazoles as kinase inhibitors
CA2796311A1 (en) 2010-04-14 2011-10-20 Incyte Corporation Fused derivatives as pi3k.delta. inhibitors
US8877707B2 (en) 2010-05-24 2014-11-04 Presidio Pharmaceuticals, Inc. Inhibitors of HCV NS5A
EP2627650A2 (en) 2010-05-26 2013-08-21 Merck Sharp & Dohme Corp. N-phenyl imidazole carboxamide inhibitors of 3-phosphoinositide-dependent protein kinase-1
WO2011163195A1 (en) 2010-06-21 2011-12-29 Incyte Corporation Fused pyrrole derivatives as pi3k inhibitors
WO2012051410A2 (en) 2010-10-13 2012-04-19 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
WO2012068440A1 (en) 2010-11-19 2012-05-24 Incyte Corporation Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as jak inhibitors
AR083933A1 (es) 2010-11-19 2013-04-10 Incyte Corp Derivados de pirrolopiridina y pirrolopirimidina sustituidos con ciclobutilo como inhibidores de jak
US8729102B2 (en) 2010-11-30 2014-05-20 Takeda Pharmaceutical Company Limited Bicyclic compound
WO2012087881A1 (en) 2010-12-20 2012-06-28 Incyte Corporation N-(1-(substituted-phenyl)ethyl)-9h-purin-6-amines as pi3k inhibitors
US9108984B2 (en) 2011-03-14 2015-08-18 Incyte Corporation Substituted diamino-pyrimidine and diamino-pyridine derivatives as PI3K inhibitors
US8673905B2 (en) 2011-03-17 2014-03-18 Hoffmann-La Roche Inc. Imidazo pyrazines
US9126948B2 (en) 2011-03-25 2015-09-08 Incyte Holdings Corporation Pyrimidine-4,6-diamine derivatives as PI3K inhibitors
WO2012143796A2 (en) * 2011-04-21 2012-10-26 Institut Pasteur Korea Anti-inflammation compounds
WO2012170867A1 (en) 2011-06-09 2012-12-13 Rhizen Pharmaceuticals Sa Novel compounds as modulators of gpr-119
EP2721028B1 (en) 2011-06-20 2015-11-04 Incyte Corporation Azetidinyl phenyl, pyridyl or pyrazinyl carboxamide derivatives as jak inhibitors
TW201313721A (zh) 2011-08-18 2013-04-01 Incyte Corp 作為jak抑制劑之環己基氮雜環丁烷衍生物
RS55737B1 (sr) 2011-09-02 2017-07-31 Incyte Holdings Corp Heterociklilamini kao inhibitori pi3k
US9212169B2 (en) 2012-03-01 2015-12-15 Hyogo College Of Medicine Benzimidazole derivative and use thereof
AR090548A1 (es) 2012-04-02 2014-11-19 Incyte Corp Azaheterociclobencilaminas biciclicas como inhibidores de pi3k
US8940742B2 (en) * 2012-04-10 2015-01-27 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9193733B2 (en) 2012-05-18 2015-11-24 Incyte Holdings Corporation Piperidinylcyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as JAK inhibitors
WO2013180193A1 (ja) 2012-05-31 2013-12-05 住友化学株式会社 縮合複素環化合物
JP6259823B2 (ja) 2012-07-13 2018-01-10 ユーシービー バイオファルマ エスピーアールエル Tnf活性の調節物質としてのイミダゾピリジン誘導体
NZ707495A (en) 2012-11-01 2019-01-25 Incyte Holdings Corp Tricyclic fused thiophene derivatives as jak inhibitors
TWI736135B (zh) 2013-03-01 2021-08-11 美商英塞特控股公司 吡唑并嘧啶衍生物治療PI3Kδ相關病症之用途
WO2014149207A2 (en) 2013-03-15 2014-09-25 Dow Agrosciences Llc Benzimidazole-based insecticidal compositions and related methods
US9656994B2 (en) 2013-03-22 2017-05-23 The Scripps Research Institute Substituted benzimidazoles as nociceptin receptor modulators
WO2014182954A1 (en) 2013-05-08 2014-11-13 Colorado Seminary, Which Owns And Operates The University Of Denver Antibiotic and anti-parasitic agents that modulate class ii fructose 1,6-bisphosphate aldolase
TR201905814T4 (tr) 2013-05-17 2019-05-21 Incyte Corp Jak inhibitörü olarak bipirazol tuzu.
SG11201600108PA (en) 2013-07-17 2016-02-26 Otsuka Pharma Co Ltd Cyanotriazole compounds
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
KR20220066179A (ko) 2014-04-08 2022-05-23 인사이트 코포레이션 Jak 및 pi3k 억제제 조합에 의한 b-세포 악성종양의 치료
EP2930048A1 (en) 2014-04-10 2015-10-14 Johnson Controls Automotive Electronics SAS Head up display projecting visual information onto a screen
WO2015191677A1 (en) 2014-06-11 2015-12-17 Incyte Corporation Bicyclic heteroarylaminoalkyl phenyl derivatives as pi3k inhibitors
MX2017003464A (es) 2014-09-16 2017-07-13 Celgene Quanticel Res Inc Inhibidores de histona desmetilasa.
WO2016054491A1 (en) * 2014-10-03 2016-04-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
MA40933A (fr) 2014-11-11 2017-09-19 Piqur Therapeutics Ag Difluorométhyl-aminopyridines et difluorométhyl-aminopyrimidines
WO2016130501A1 (en) 2015-02-09 2016-08-18 Incyte Corporation Aza-heteroaryl compounds as pi3k-gamma inhibitors
TWI824309B (zh) 2015-11-06 2023-12-01 美商英塞特公司 作為PI3K-γ抑制劑之雜環化合物
US20170190689A1 (en) 2016-01-05 2017-07-06 Incyte Corporation Pyridine and pyridimine compounds as pi3k-gamma inhibitors
WO2017223414A1 (en) 2016-06-24 2017-12-28 Incyte Corporation HETEROCYCLIC COMPOUNDS AS PI3K-γ INHIBITORS

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
EP3371190A1 (en) 2018-09-12
US20230009843A1 (en) 2023-01-12
TW202204362A (zh) 2022-02-01
MA43169B1 (fr) 2022-05-31
PL3371190T3 (pl) 2022-11-07
US11091491B2 (en) 2021-08-17
ES2915550T3 (es) 2022-06-23
JP6854817B2 (ja) 2021-04-07
RS63359B1 (sr) 2022-07-29
US11773102B2 (en) 2023-10-03
JP2018532760A (ja) 2018-11-08
EP4086259A1 (en) 2022-11-09
DK3371190T3 (da) 2022-05-23
US20200031837A1 (en) 2020-01-30
WO2017079519A1 (en) 2017-05-11
HUE059324T2 (hu) 2022-11-28
US20190062336A1 (en) 2019-02-28
TWI824309B (zh) 2023-12-01
MA43169A (fr) 2018-09-12
HRP20220599T1 (hr) 2022-06-24
PT3371190T (pt) 2022-07-08
AR106595A1 (es) 2018-01-31
US10065963B2 (en) 2018-09-04
LT3371190T (lt) 2022-08-10
US20170129899A1 (en) 2017-05-11
US10472368B2 (en) 2019-11-12
TW201726679A (zh) 2017-08-01
TWI744256B (zh) 2021-11-01
SI3371190T1 (sl) 2022-08-31
MD3371190T2 (ro) 2022-09-30

Similar Documents

Publication Publication Date Title
US11773102B2 (en) Heterocyclic compounds as PI3K-γ inhibitors
US10975088B2 (en) Imidazo[2,1-f][1,2,4]triazine compounds as pi3k-y inhibitors
US11053215B2 (en) Heterocyclic compounds useful as Pim kinase inhibitors
US10519163B2 (en) Pyrrolopyrimidine derivatives as TAM inhibitors
EP3400221B1 (en) Pyrazol / imidazol substituted pyridines as pi3k-gamma inhibitors
US20230338389A1 (en) Bicyclic heterocycles as fgfr inhibitors
EP3818063A1 (en) Fused pyrazine derivatives as a2a / a2b inhibitors
KR20130116358A (ko) Pi3 키나아제 억제제로서 헤테로사이클릭 화합물
KR20200084905A (ko) Pi3k 억제제로서 헤테로시클릴아민
JP7399968B2 (ja) Alk2及び/またはfgfr調節剤としてのピラゾロ[4,3-d]ピリミジン化合物
KR20210116488A (ko) 액티빈 수용체-유사 키나제-2의 억제제로서의 이미다조피리다진 및 이미다조피리딘 화합물
US11999740B2 (en) Fused pyrazine derivatives as A2A / A2B inhibitors

Legal Events

Date Code Title Description
REG Reference to a national code

Ref country code: HR

Ref legal event code: TUEP

Ref document number: P20220599

Country of ref document: HR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20180601

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1259750

Country of ref document: HK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20200327

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20211005

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602016070555

Country of ref document: DE

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1479112

Country of ref document: AT

Kind code of ref document: T

Effective date: 20220415

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20220518

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: MA

Ref legal event code: VAGR

Ref document number: 43169

Country of ref document: MA

Kind code of ref document: B1

REG Reference to a national code

Ref country code: FI

Ref legal event code: FGE

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2915550

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20220623

REG Reference to a national code

Ref country code: HR

Ref legal event code: T1PR

Ref document number: P20220599

Country of ref document: HR

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Ref document number: 3371190

Country of ref document: PT

Date of ref document: 20220708

Kind code of ref document: T

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20220630

REG Reference to a national code

Ref country code: EE

Ref legal event code: FG4A

Ref document number: E022435

Country of ref document: EE

Effective date: 20220621

Ref country code: NO

Ref legal event code: T2

Effective date: 20220330

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20220401200

Country of ref document: GR

Effective date: 20220707

REG Reference to a national code

Ref country code: MD

Ref legal event code: VAGR

Ref document number: 3371190

Country of ref document: MD

Date of ref document: 20220930

Kind code of ref document: T2

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20220599

Country of ref document: HR

Payment date: 20221102

Year of fee payment: 7

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E059324

Country of ref document: HU

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602016070555

Country of ref document: DE

VSFP Annual fee paid to validation state [announced via postgrant information from national office to epo]

Ref country code: MD

Payment date: 20221110

Year of fee payment: 7

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20230103

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 1479112

Country of ref document: AT

Kind code of ref document: T

Effective date: 20220330

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230526

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: AL

Payment date: 20221120

Year of fee payment: 7

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20220599

Country of ref document: HR

Payment date: 20231019

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20231126

Year of fee payment: 8

Ref country code: LU

Payment date: 20231127

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SK

Payment date: 20231018

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GR

Payment date: 20231129

Year of fee payment: 8

Ref country code: GB

Payment date: 20231127

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MC

Payment date: 20231020

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20231201

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IS

Payment date: 20231025

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20231024

Year of fee payment: 8

Ref country code: SM

Payment date: 20231027

Year of fee payment: 8

Ref country code: SI

Payment date: 20231019

Year of fee payment: 8

Ref country code: SE

Payment date: 20231127

Year of fee payment: 8

Ref country code: RS

Payment date: 20231019

Year of fee payment: 8

Ref country code: RO

Payment date: 20231020

Year of fee payment: 8

Ref country code: PT

Payment date: 20231020

Year of fee payment: 8

Ref country code: NO

Payment date: 20231129

Year of fee payment: 8

Ref country code: MT

Payment date: 20231018

Year of fee payment: 8

Ref country code: LV

Payment date: 20231024

Year of fee payment: 8

Ref country code: LT

Payment date: 20231018

Year of fee payment: 8

Ref country code: IT

Payment date: 20231122

Year of fee payment: 8

Ref country code: IE

Payment date: 20231127

Year of fee payment: 8

Ref country code: HU

Payment date: 20231031

Year of fee payment: 8

Ref country code: HR

Payment date: 20231019

Year of fee payment: 8

Ref country code: FR

Payment date: 20231127

Year of fee payment: 8

Ref country code: FI

Payment date: 20231127

Year of fee payment: 8

Ref country code: EE

Payment date: 20231023

Year of fee payment: 8

Ref country code: DK

Payment date: 20231127

Year of fee payment: 8

Ref country code: DE

Payment date: 20231129

Year of fee payment: 8

Ref country code: CZ

Payment date: 20231025

Year of fee payment: 8

Ref country code: CY

Payment date: 20231019

Year of fee payment: 8

Ref country code: CH

Payment date: 20231201

Year of fee payment: 8

Ref country code: BG

Payment date: 20231121

Year of fee payment: 8

Ref country code: AT

Payment date: 20231019

Year of fee payment: 8

VSFP Annual fee paid to validation state [announced via postgrant information from national office to epo]

Ref country code: MD

Payment date: 20231025

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PL

Payment date: 20231020

Year of fee payment: 8

Ref country code: BE

Payment date: 20231127

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: MK

Payment date: 20231019

Year of fee payment: 8